1
|
Kadeh H, Baranzehi T, Mollaali M, Maserat N, Shahraki MJ, Kordi-Tamandani DM. Correlation between JAK2, STAT3, and CTLA4 Relative Gene Expressions and Oral Squamous Cell Carcinoma. JOURNAL OF DENTISTRY (SHIRAZ, IRAN) 2025; 26:25-32. [PMID: 40092547 PMCID: PMC11909409 DOI: 10.30476/dentjods.2024.100237.2205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/05/2023] [Accepted: 02/24/2024] [Indexed: 03/19/2025]
Abstract
Statement of the Problem Oral squamous cell carcinoma (OSCC) is the eighth leading cause of cancer-related death worldwide. JAK2 and STAT3 primarily influence intrinsic tumor cell behavior, and CTLA4 impacts the interplay between the tumor and the host immune system in the context of cancers. There is scarce information regarding the involvement and roles of JAK2, STAT3, and CTLA4 genes in OSCC; however, the molecular mechanisms are still unclear. Purpose This study examined the relationship between JAK2, STAT3, and CTLA4 gene expression levels and OSCC in a group of patients in the southeast of Iran. Materials and Method This cross-sectional study was conducted in which the relative gene expression levels of JAK2, STAT3, and CTLA4 were compared between 23 oral paraffin tissue blocks collected from OSCC patients and 20 fresh gingival tissues collected from healthy individuals. The Real-Time quantitative PCR (RT-qPCR) assay was employed to assess relative gene expression levels. SPSS 27 was employed to perform statistical analyses. Results Significant differences were found between OSCC patients and healthy individuals concerning gene expression levels of JAK2 (2.4-fold, p< 0.0001), STAT3 (2.32-fold, p< 0.0001), and CTLA4 (4.09-fold, p< 0.0001). Additionally, there were significant positive correlations among JAK2-STAT3 (ρ= 0.667, p< 0.001), JAK2-CTLA4 (ρ= 0.771, p< 0.001), and STAT3-CTLA4 (ρ= 0.635, p= 0.001) co-expressions. Moreover, gender, age groups, and tumor locations did not significantly correlate with the expression levels of these genes (p> 0.05). Nevertheless, significant differences occurred between histopathological grades and the gene expression levels of JAK2 (p< 0.001), STAT3 (p= 0.001), and CTLA4 (p< 0.001). Conclusion The overexpression of JAK2, STAT3, and CTLA4 can be considered triggers for OSCC development. It may be beneficial to conduct future research on OSCC by considering downstream genes involved in the JAK2/STAT3/CTLA4 axis.
Collapse
Affiliation(s)
- Hamideh Kadeh
- Dept. Oral and Maxillofacial Pathology, Oral and Dental Disease Research Center, Faculty of Dentistry, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Tayebeh Baranzehi
- Dept. of Biology, Faculty of Science, University of Sistan and Baluchestan, Zahedan, Iran
| | - Milad Mollaali
- Dept. of Biology, Faculty of Science, University of Sistan and Baluchestan, Zahedan, Iran
| | - Neda Maserat
- Dept. of Biology, Faculty of Science, University of Sistan and Baluchestan, Zahedan, Iran
| | | | | |
Collapse
|
2
|
Chen JY, Li YF, Zhou Z, Jiang XM, Bi X, Yang MF, Zhao B. De novo mutations promote inflammation in children with STAT3 gain-of-function syndrome by affecting IL-1β expression. Int Immunopharmacol 2024; 140:112755. [PMID: 39098225 DOI: 10.1016/j.intimp.2024.112755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/09/2024] [Accepted: 07/22/2024] [Indexed: 08/06/2024]
Abstract
STAT3 gain-of-function syndrome, characterized by early-onset autoimmunity and primary immune regulatory disorder, remains poorly understood in terms of its immunological mechanisms. We employed whole-genome sequencing of familial trios to elucidate the pivotal role of de novo mutations in genetic diseases. We identified 37 high-risk pathogenic loci affecting 23 genes, including a novel STAT3 c.508G>A mutation. We also observed significant down-regulation of pathogenic genes in affected individuals, potentially associated with inflammatory responses regulated by PTPN14 via miR378c. These findings enhance our understanding of the pathogenesis of STAT3 gain-of-function syndrome and suggest potential therapeutic strategies. Notably, combined JAK inhibitors and IL-6R antagonists may offer promising treatment avenues for mitigating the severity of STAT3 gain-of-function syndrome.
Collapse
Affiliation(s)
- Ji-Yu Chen
- Department of Nephrology & Rheumatology, Kunming Children's Hospital, Kunming 650228, Yunnan, China
| | - Yan-Fang Li
- Department of Nephrology & Rheumatology, Kunming Children's Hospital, Kunming 650228, Yunnan, China
| | - Zhu Zhou
- Department of Nephrology, First Affiliated Hospital of Kunming Medical University, Yunnan Clinical Medical Research Center of Chronic Kidney Disease, Kunming 650032, Yunnan, China
| | - Xue-Mei Jiang
- Department of Nephrology & Rheumatology, Kunming Children's Hospital, Kunming 650228, Yunnan, China
| | - Xin Bi
- Department of Nephrology & Rheumatology, Kunming Children's Hospital, Kunming 650228, Yunnan, China
| | - Mi-Feng Yang
- Department of Nephrology & Rheumatology, Kunming Children's Hospital, Kunming 650228, Yunnan, China
| | - Bo Zhao
- Department of Nephrology & Rheumatology, Kunming Children's Hospital, Kunming 650228, Yunnan, China.
| |
Collapse
|
3
|
Hussein Zaki A, Haiying B, Mohany M, Al-Rejaie SS, Abugammie B. The effect mechanism of ergosterol from the nutritional mushroom Leucocalocybe mongolica in breast cancer cells: Protein expression modulation and metabolomic profiling using UHPLC-ESI-Q. Saudi Pharm J 2024; 32:102045. [PMID: 38571766 PMCID: PMC10988126 DOI: 10.1016/j.jsps.2024.102045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
The ergosterol from mushrooms has gained significant ethnopharmacological importance in various cultures, including China, Japan, and Europe. This compound has been found to possess immune-boosting and anti-inflammatory properties, making it useful in the treatment of immune disorders. In this study, we focused on investigating the potential anticancer properties of ergosterol isolated from the edible mushroom Leucocalocybe mongolica in breast cancer cell lines. The ergosterol was purified and identified using advanced analytical techniques such as ESI-MS and NMR. We conducted cell proliferation assays on 4 T1 breast cancer cells to assess the cytotoxic effects of ergosterol. Furthermore, we analyzed the transcription levels of BAX, caspase-7, BCL-2, STAT-3, and PARP proteins using real-time PCR and Western blot analysis. Additionally, we employed non-targeted ultra-high-performance liquid chromatography and high-resolution mass spectrometry (UPLC-MS/MS) to study the potential mechanisms underlying the anticancer effects of ergosterol at the metabolomics level. The results demonstrated a significant reduction in cell viability and the induction of apoptosis upon treatment with ergosterol, especially at higher concentrations (P < 0.05). Moreover, ergosterol affected the expression of cancer-related genes, upregulating pro-apoptotic proteins such as BAX, caspase-7, and PARP, while downregulating the anti-apoptotic proteins BCL-2 and STAT-3 (P < 0.05). Western blot analysis confirmed these findings and provided further evidence of ergosterol's role in inducing apoptosis. Metabolomics analysis revealed substantial changes in pathways related to amino acid, antioxidant, and carbohydrate metabolism. In conclusion, our study demonstrates that ergosterol exhibits anticancer effects by inducing apoptosis and modulating metabolic pathways in breast cancer cells.
Collapse
Affiliation(s)
- Asmaa Hussein Zaki
- Key Laboratory of Edible Fungi Resources and Utilization, Ministry of Agriculture and Rural Affairs, Jilin Agricultural University, Changchun 130118, Jilin, China
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun 130118, Jilin, China
- Departments of Agricultural Chemistry, Faculty of Agriculture, Minia University, El-Minia 61519, Egypt
| | - Bao Haiying
- Key Laboratory of Edible Fungi Resources and Utilization, Ministry of Agriculture and Rural Affairs, Jilin Agricultural University, Changchun 130118, Jilin, China
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Mohamed Mohany
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 55760, Riyadh 11451, Saudi Arabia
| | - Salim S. Al-Rejaie
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 55760, Riyadh 11451, Saudi Arabia
| | - Bahaa Abugammie
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, China
| |
Collapse
|
4
|
Bancet A, Frem R, Jeanneret F, Mularoni A, Bazelle P, Roelants C, Delcros JG, Guichou JF, Pillet C, Coste I, Renno T, Battail C, Cochet C, Lomberget T, Filhol O, Krimm I. Cancer selective cell death induction by a bivalent CK2 inhibitor targeting the ATP site and the allosteric αD pocket. iScience 2024; 27:108903. [PMID: 38318383 PMCID: PMC10838953 DOI: 10.1016/j.isci.2024.108903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/25/2023] [Accepted: 01/09/2024] [Indexed: 02/07/2024] Open
Abstract
Although the involvement of protein kinase CK2 in cancer is well-documented, there is a need for selective CK2 inhibitors suitable for investigating CK2 specific roles in cancer-related biological pathways and further exploring its therapeutic potential. Here, we report the discovery of AB668, an outstanding selective inhibitor that binds CK2 through a bivalent mode, interacting both at the ATP site and an allosteric αD pocket unique to CK2. Using caspase activation assay, live-cell imaging, and transcriptomic analysis, we have compared the effects of this bivalent inhibitor to representative ATP-competitive inhibitors, CX-4945, and SGC-CK2-1. Our results show that in contrast to CX-4945 or SGC-CK2-1, AB668, by targeting the CK2 αD pocket, has a distinct mechanism of action regarding its anti-cancer activity, inducing apoptotic cell death in several cancer cell lines and stimulating distinct biological pathways in renal cell carcinoma.
Collapse
Affiliation(s)
- Alexandre Bancet
- University Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Institut Convergence Plascan, Team « Small Molecules for Biological Targets », 69373 Lyon, France
- Kairos Discovery SAS, 36 Rue Jeanne d’Arc, 69003 Lyon, France
| | - Rita Frem
- University Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Institut Convergence Plascan, Team « Targeting Non-canonical Protein Functions in Cancer », 69373 Lyon, France
| | - Florian Jeanneret
- Université Grenoble Alpes, IRIG, Laboratoire Biosciences et Bioingénierie pour la Santé, UA 13 INSERM-CEA-UGA, 38000 Grenoble, France
| | - Angélique Mularoni
- University Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Institut Convergence Plascan, Team « Small Molecules for Biological Targets », 69373 Lyon, France
| | - Pauline Bazelle
- Université Grenoble Alpes, IRIG, Laboratoire Biosciences et Bioingénierie pour la Santé, UA 13 INSERM-CEA-UGA, 38000 Grenoble, France
| | - Caroline Roelants
- University Grenoble Alpes, INSERM 1292, CEA, UMR Biosanté, 38000 Grenoble, France
| | - Jean-Guy Delcros
- University Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Institut Convergence Plascan, Team « Small Molecules for Biological Targets », 69373 Lyon, France
| | - Jean-François Guichou
- Centre de Biologie Structurale, CNRS, INSERM, University Montpellier, 34090 Montpellier, France
| | - Catherine Pillet
- University Grenoble Alpes, INSERM 1292, CEA, UMR Biosanté, 38000 Grenoble, France
| | - Isabelle Coste
- University Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Institut Convergence Plascan, Team « Targeting Non-canonical Protein Functions in Cancer », 69373 Lyon, France
| | - Toufic Renno
- University Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Institut Convergence Plascan, Team « Targeting Non-canonical Protein Functions in Cancer », 69373 Lyon, France
| | - Christophe Battail
- Université Grenoble Alpes, IRIG, Laboratoire Biosciences et Bioingénierie pour la Santé, UA 13 INSERM-CEA-UGA, 38000 Grenoble, France
| | - Claude Cochet
- University Grenoble Alpes, INSERM 1292, CEA, UMR Biosanté, 38000 Grenoble, France
| | - Thierry Lomberget
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), COSSBA Team, Faculté de Pharmacie-ISPB, 8 Avenue Rockefeller, 69373 Lyon Cedex 08, France
| | - Odile Filhol
- University Grenoble Alpes, INSERM 1292, CEA, UMR Biosanté, 38000 Grenoble, France
| | - Isabelle Krimm
- University Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Institut Convergence Plascan, Team « Small Molecules for Biological Targets », 69373 Lyon, France
| |
Collapse
|
5
|
Bose M, Sanders A, Handa A, Vora A, Cardona MR, Brouwer C, Mukherjee P. Molecular crosstalk between MUC1 and STAT3 influences the anti-proliferative effect of Napabucasin in epithelial cancers. Sci Rep 2024; 14:3178. [PMID: 38326371 PMCID: PMC10850135 DOI: 10.1038/s41598-024-53549-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 02/01/2024] [Indexed: 02/09/2024] Open
Abstract
MUC1 is a transmembrane glycoprotein that is overexpressed and aberrantly glycosylated in epithelial cancers. The cytoplasmic tail of MUC1 (MUC1 CT) aids in tumorigenesis by upregulating the expression of multiple oncogenes. Signal transducer and activator of transcription 3 (STAT3) plays a crucial role in several cellular processes and is aberrantly activated in many cancers. In this study, we focus on recent evidence suggesting that STAT3 and MUC1 regulate each other's expression in cancer cells in an auto-inductive loop and found that their interaction plays a prominent role in mediating epithelial-to-mesenchymal transition (EMT) and drug resistance. The STAT3 inhibitor Napabucasin was in clinical trials but was discontinued due to futility. We found that higher expression of MUC1 increased the sensitivity of cancer cells to Napabucasin. Therefore, high-MUC1 tumors may have a better outcome to Napabucasin therapy. We report how MUC1 regulates STAT3 activity and provide a new perspective on repurposing the STAT3-inhibitor Napabucasin to improve clinical outcome of epithelial cancer treatment.
Collapse
Affiliation(s)
- Mukulika Bose
- Department of Biological Sciences, UNC Charlotte, Charlotte, NC, 28223, USA.
| | - Alexa Sanders
- Department of Bioinformatics, UNC Charlotte, Charlotte, NC, 28223, USA
| | - Aashna Handa
- Department of Biological Sciences, UNC Charlotte, Charlotte, NC, 28223, USA
| | - Aabha Vora
- Department of Biological Sciences, UNC Charlotte, Charlotte, NC, 28223, USA
| | - Manuel R Cardona
- Department of Biological Sciences, UNC Charlotte, Charlotte, NC, 28223, USA
| | - Cory Brouwer
- Department of Bioinformatics, UNC Charlotte, Charlotte, NC, 28223, USA
| | - Pinku Mukherjee
- Department of Biological Sciences, UNC Charlotte, Charlotte, NC, 28223, USA.
| |
Collapse
|
6
|
JIN T, ZHOU Q, SHEN J, ZHANG Z, LIAN X. Caffeic acid 3,4-dihydroxyphenethyl ester prevents colorectal cancer through inhibition of multiple cancer-promoting signal pathways in 1,2-Dimethylhydrazine/dextran sodium sulphate mouse model. J TRADIT CHIN MED 2024; 44:70-77. [PMID: 38213241 PMCID: PMC10774738 DOI: 10.19852/j.cnki.jtcm.20231204.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/17/2022] [Indexed: 01/13/2024]
Abstract
OBJECTIVE To elucidate the potential feature and mechanism of the caffeic acid 3,4-dihydroxyphenethyl ester (CADPE) molecule, which can prevent colorectal cancer (CRC) in the 1,2-Dimethylhydrazine (DMH)/dextran sodium sulphate (DSS)-induced mouse model. METHODS Institute of cancer research (ICR) male mice were injected with 20 mg/kg DMH for a week. After that, 2% DSS was administered in the drinking water for another 7 d. The CADPE treatment was given to the DMH/DSS induced male mice at three different periods until their sacrifice. Histopathological examination was used for observing the CRC development at colonic mucosa. Immunohistochemistry (IHC), blood cells smearing and crypt damage scoring methods were used for investigating the anti-inflammation feature of CADPE related to CRC. The reversing targets searching method was applied with artificial intelligence (AI), computer-aided drug designing (CADD) and Ingenuity Pathway Analysis (IPA) techniques for predicting the potential targets and mechanism of CADPE highly related to CRC. RESULTS The data indicated that CADPE inhibited CRC tumor development in the colitis-associated DMH/DSS induced mouse model after giving the early treatment. CADPE also impeded the acute inflammation by decreasing the infiltration of neutrophils significantly during the initial stage of CRC development. Finally, our data showed that CADPE prevented CRC by blocking active sites of three pivotal protein targets including epidermal growth factor receptor (EGFR), extracellular signal-regulated kinase (ERK) and mammalian target of rapamycin (mTOR) in two major cancer development pathways. CONCLUSIONS CADPE effectively prevented CRC at early stage of tumor germination in the DMH/DSS mouse model highly likely due to its anti-acute inflammation characteristic and the ability of blocking EGFR, ERK and mTOR activities in two highly related CRC developing pathways.
Collapse
Affiliation(s)
- Tao JIN
- 1 College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qian ZHOU
- 1 College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jichen SHEN
- 2 Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhizhong ZHANG
- 3 Ocean College, Zhoushan Campus of Zhejiang University, Zhoushan 316021, China
| | - Xiaoyuan LIAN
- 4 College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
7
|
Tong Y, Cao Y, Jin T, Huang Z, He Q, Mao M. Role of Interleukin-1 family in bone metastasis of prostate cancer. Front Oncol 2022; 12:951167. [PMID: 36237303 PMCID: PMC9552844 DOI: 10.3389/fonc.2022.951167] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/12/2022] [Indexed: 11/28/2022] Open
Abstract
Prostate cancer (PCa) is one of the most fatal diseases in male patients with high bone metastatic potential. Bone metastasis severely shortens overall survival and brings skeletal-related events (SREs) which reduces the life quality of patients, and this situation is currently regarded as irreversible and incurable. The progression and metastasis of PCa are found to be closely associated with inflammatory cytokines and chemokines. As pivotal members of inflammatory cytokines, Interleukin-1 (IL-1) family plays a crucial role in this process. Elevated expression of IL-1 family was detected in PCa patients with bone metastasis, and accumulating evidences proved that IL-1 family could exert vital effects on the progression and bone metastasis of many cancers, while some members have dual effects. In this review, we discuss the role of IL-1 family in the bone metastasis of PCa. Furthermore, we demonstrate that many members of IL-1 family could act as pivotal biomarkers to predict the clinical stage and prognosis of PCa patients. More importantly, we have elucidated the role of IL-1 family in the bone metastasis of PCa, which could provide potential targets for the treatment of PCa bone metastasis and probable directions for future research.
Collapse
Affiliation(s)
- Yuanhao Tong
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Yinghao Cao
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianzhe Jin
- Department of Gynecologic Oncology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhengwei Huang
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Qinyuan He
- Organization Department, Suzhou Traditional Chinese Medicine Hospital, Suzhou, China
| | - Min Mao
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Min Mao,
| |
Collapse
|
8
|
Xu J, Kim H, Dong J, Chen H, Xu J, Ma R, Zhou M, Wang T, Shen Q, Zhou J. Structure-activity relationship studies on O-alkylamino-tethered salicylamide derivatives with various amino acid linkers as potent anticancer agents. Eur J Med Chem 2022; 234:114229. [PMID: 35334447 PMCID: PMC9040195 DOI: 10.1016/j.ejmech.2022.114229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/09/2022] [Accepted: 02/22/2022] [Indexed: 11/15/2022]
Abstract
In our continued SAR study efforts, a series of O-alkylamino-tethered salicylamide derivatives with various amino acid linkers has been designed, synthesized, and biologically evaluated as potent anticancer agents. Five selected compounds with different representative chemical structures were found to show broad anti-proliferative activities, effective against all tested ER-positive breast cancer (BC) and triple-negative breast cancer (TNBC) cell lines with low micromolar IC50 values. Among these compounds, compound 9a (JMX0293) maintained good potency against MDA-MB-231 cell line (IC50 = 3.38 ± 0.37 μM) while exhibiting very low toxicity against human non-tumorigenic breast epithelial cell line MCF-10A (IC50 > 60 μM). Further mechanistic studies showed that compound 9a could inhibit STAT3 phosphorylation and contribute to apoptosis in TNBC MDA-MB-231 cells. More importantly, compound 9a significantly suppressed MDA-MB-231 xenograft tumor growth in vivo without significant toxicity, indicating its great potential as a promising anticancer drug candidate for further clinical development.
Collapse
Affiliation(s)
- Jimin Xu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, United States
| | - Hyejin Kim
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, United States
| | - Jiabin Dong
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, United States
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, United States
| | - Junhai Xu
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, United States
| | - Ruixia Ma
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, United States
| | - Mingxiang Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, United States
| | - Tianzhi Wang
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, 77555, United States
| | - Qiang Shen
- Department of Interdisciplinary Oncology, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, United States.
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, United States; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, 77555, United States.
| |
Collapse
|
9
|
Targeted Protein Profiling of In Vivo NIPP-Treated Tissues Using DigiWest Technology. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app112311238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Non-invasive physical plasma (NIPP) is a novel therapeutic tool, currently being evaluated for the treatment of cancer and precancerous lesions in gynecology and other disciplines. Additionally, patients with cervical intraepithelial neoplasia (CIN) may benefit from NIPP treatment due to its non-invasive, side-effect-free, and tissue-sparing character. However, the molecular impact of in vivo NIPP treatment needs to be further investigated. For this purpose, usually only very small tissue biopsies are available after NIPP treatment. Here, we adapted DigiWest technology, a high-throughput bead-based Western blot, for the analysis of formalin-fixed paraffin-embedded (FFPE) cervical punch biopsies with a minimal sample amount. We investigated the molecular effects of NIPP treatment directly after (0 h) and 24 h after in vivo application. Results were compared to in vitro NIPP-treated human malignant cervical cells. NIPP effects were primarily based on an inhibitory impact on the cell cycle and cell growth factors. DigiWest technology was suitable for detailed protein profiling of small, primary FFPE biopsies.
Collapse
|
10
|
Boersma B, Jiskoot W, Lowe P, Bourquin C. The interleukin-1 cytokine family members: Role in cancer pathogenesis and potential therapeutic applications in cancer immunotherapy. Cytokine Growth Factor Rev 2021; 62:1-14. [PMID: 34620560 DOI: 10.1016/j.cytogfr.2021.09.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/20/2021] [Indexed: 02/06/2023]
Abstract
The interleukin-1 (IL-1) family is one of the first described cytokine families and consists of eight cytokines (IL-1β, IL-1α, IL-18, IL-33, IL-36α, IL-36β, IL-36γ and IL-37) and three receptor antagonists (IL-1Ra, IL-36Ra and IL-38). The family members are known to play an essential role in inflammation. The importance of inflammation in cancer has been well established in the past decades. This review sets out to give an overview of the role of each IL-1 family member in cancer pathogenesis and show their potential as potential anticancer drug candidates. First, the molecular structure is described. Next, both the pro- and anti-tumoral properties are highlighted. Additionally, a critical interpretation of current literature is given. To conclude, the IL-1 family is a toolbox with a collection of powerful tools that can be considered as potential drugs or drug targets.
Collapse
Affiliation(s)
- Bart Boersma
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland; School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland.
| | - Wim Jiskoot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| | - Peter Lowe
- Department of Biomolecule Generation and Optimization, Institut de Recherche Pierre Fabre, Centre d'Immunologie Pierre Fabre, Saint-Julien-en-Genevois, France.
| | - Carole Bourquin
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland; School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; Department of Anesthesiology, Pharmacology and Intensive Care, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
11
|
Crucial Role of Reactive Oxygen Species (ROS) for the Proapoptotic Effects of Indirubin Derivatives in Cutaneous SCC Cells. Antioxidants (Basel) 2021; 10:antiox10101514. [PMID: 34679649 PMCID: PMC8532942 DOI: 10.3390/antiox10101514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/31/2021] [Accepted: 09/13/2021] [Indexed: 01/04/2023] Open
Abstract
Efficient drugs are needed for countering the worldwide high incidence of cutaneous squamous cell carcinoma (cSCC) and actinic keratosis. Indirubin derivatives represent promising candidates, but their effects in cSCC cells have not been reported before. Here, we investigated the efficacy of three indirubin derivatives (DKP-071, -073 and -184) in four cSCC cell lines. High efficacy was seen in SCL-I, SCL-II, SCC-12 and SCC-13, resulting in up to 80% loss of cell proliferation, 60% loss of cell viability and 30% induced apoptosis (10 µM). Apoptosis was further enhanced in combinations with TNF-related apoptosis-inducing ligand (TRAIL). Induction of reactive oxygen species (ROS) appeared as critical for these effects. Thus, antioxidative pretreatment completely abolished apoptosis as well as restored cell proliferation and viability. Concerning the pathways, complete activation of caspases cascades (caspases-3, -4, -6, -7, -8 and -9), loss of mitochondrial membrane potential, activation of proapoptotic PKCδ (protein kinase C delta), inhibition of STAT3 (signal transducer and activator of transcription 3), downregulation of antiapoptotic XIAP (X-linked inhibitor of apoptosis protein) and survivin as well as upregulation of the proapoptotic Bcl-2 protein Puma and the cell cycle inhibitor p21 were obtained. Importantly, all activation steps were prevented by antioxidants, thus proving ROS as a master regulator of indirubins' antitumor effects. ROS induction presently develops as an important issue in anticancer therapy.
Collapse
|
12
|
Effects of Fatty Acid Oxidation and Its Regulation on Dendritic Cell-Mediated Immune Responses in Allergies: An Immunometabolism Perspective. J Immunol Res 2021; 2021:7483865. [PMID: 34423053 PMCID: PMC8376428 DOI: 10.1155/2021/7483865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 07/08/2021] [Accepted: 07/27/2021] [Indexed: 12/25/2022] Open
Abstract
Type 1 allergies, involve a complex interaction between dendritic cells and other immune cells, are pathological type 2 inflammatory immune responses against harmless allergens. Activated dendritic cells undergo extensive phenotypic and functional changes to exert their functions. The activation, differentiation, proliferation, migration, and mounting of effector reactions require metabolic reprogramming. Dendritic cells are important upstream mediators of allergic responses and are therefore an important effector of allergies. Hence, a better understanding of the underlying metabolic mechanisms of functional changes that promote allergic responses of dendritic cells could improve the prevention and treatment of allergies. Metabolic changes related to dendritic cell activation have been extensively studied. This review briefly outlines the basis of fatty acid oxidation and its association with dendritic cell immune responses. The relationship between immune metabolism and effector function of dendritic cells related to allergic diseases can better explain the induction and maintenance of allergic responses. Further investigations are warranted to improve our understanding of disease pathology and enable new treatment strategies.
Collapse
|
13
|
Yue CH, Oner M, Chiu CY, Chen MC, Teng CL, Wang HY, Hsieh JT, Lai CH, Lin H. RET Regulates Human Medullary Thyroid Cancer Cell Proliferation through CDK5 and STAT3 Activation. Biomolecules 2021; 11:biom11060860. [PMID: 34207842 PMCID: PMC8229599 DOI: 10.3390/biom11060860] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
Medullary thyroid cancer (MTC) is a neuroendocrine tumor that arises from the parafollicular C-cells, which produces the hormone calcitonin. RET is a transmembrane receptor protein-tyrosine kinase, which is highly expressed in MTC. Our previous studies reported that cyclin-dependent kinase 5 (CDK5) plays a crucial role in cancer progression, including MTC. However, the role of CDK5 in GDNF-induced RET signaling in medullary thyroid cancer proliferation remains unknown. Here, we investigated RET activation and its biochemically interaction with CDK5 in GDNF-induced medullary thyroid cancer proliferation. Our results demonstrated that GDNF stimulated RET phosphorylation and thus subsequently resulted in CDK5 activation by its phosphorylation. Activated CDK5 further caused STAT3 activation by its specific phosphorylation at Ser727. Moreover, we also found that GDNF treatment enhanced ERK1/2 and EGR1 activity, which is involved in p35 activation. Interestingly, we identified for the first time that CDK5 physically interacted with RET protein in MTC. Overall, our results provide a new mechanism for medullary thyroid cancer cell proliferation, suggesting that targeting CDK5 may be a promising therapeutic candidate for human medullary thyroid cancer in the near future.
Collapse
Affiliation(s)
- Chia-Herng Yue
- Department of Surgery, Tung’s Taichung Metro Harbor Hospital, Taichung 435403, Taiwan;
| | - Muhammet Oner
- Department of Life Sciences, National Chung Hsing University, Taichung 402204, Taiwan; (M.O.); (C.-Y.C.)
| | - Chih-Yuan Chiu
- Department of Life Sciences, National Chung Hsing University, Taichung 402204, Taiwan; (M.O.); (C.-Y.C.)
| | - Mei-Chih Chen
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung 404332, Taiwan;
| | - Chieh-Lin Teng
- Division of Hematology/Medical Oncology, Department of Medicine, Taichung Veterans General Hospital, Taichung 40201, Taiwan;
- Department of Life Science, Tunghai University, Taichung 40704, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Hsin-Yi Wang
- Department of Nuclear Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung 402204, Taiwan; (M.O.); (C.-Y.C.)
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402204, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402204, Taiwan
- Correspondence: ; Tel.: +886-4-22840-416 (ext. 311); Fax: +886-4-22874-740
| |
Collapse
|
14
|
Isoorientin inhibits epithelial-to-mesenchymal properties and cancer stem-cell-like features in oral squamous cell carcinoma by blocking Wnt/β-catenin/STAT3 axis. Toxicol Appl Pharmacol 2021; 424:115581. [PMID: 34019859 DOI: 10.1016/j.taap.2021.115581] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 05/14/2021] [Accepted: 05/16/2021] [Indexed: 01/22/2023]
Abstract
Oral squamous cell carcinoma (OSCC) is among the most prevalent cancers of the head and neck. This study revealed that isoorientin attenuates OSCC cell stemness and epithelial-mesenchymal transition potential through the inhibition of JAK/signal transducer and activator of transcription 3 (STAT3) and Wnt/β-catenin signaling in cell lines. Our findings indicated that isoorientin is a potential inhibitor of β-catenin/STAT3 in vitro and in vivo. We analyzed possible synergism between isoorientin and cisplatin in OSCC. A sulforhodamine B assay, colony formation assay, tumorsphere-formation assay, and Wnt reporter activity assay were used for determining cell invasion, cell migration, drug cytotoxicity, and cell viability with potential molecular mechanisms in vitro. Isoorientin reduced the expression of p-STAT3, β-catenin, and p-GSK3 as well as downstream effectors TCF1/TCF7 and LEF1 and significantly reduced β-catenin colocalization in the nucleus. Isoorientin markedly strengthened the cytotoxic effects of cisplatin against SAS and SCC-25. Therefore, combining isoorientin and cisplatin treatments can potentially improve the anticancer effect of cisplatin. Isoorientin inhibited the tumorigenicity and growth of OSCC through the abrogation of Wnt/β-catenin/STAT3 signaling in vivo. Thus, isoorientin disrupted the β-catenin signaling pathway through the inactivation of STAT3 signaling. In conclusion, targeting OSCC-SC-mediated stemness with isoorientin to eradicate OSCC-SCs may be an effective strategy for preventing relapse and metastasis of OSCC and providing long-term survival benefits.
Collapse
|
15
|
Elhennawy MG, Abdelaleem EA, Zaki AA, Mohamed WR. Cinnamaldehyde and hesperetin attenuate TNBS-induced ulcerative colitis in rats through modulation of the JAk2/STAT3/SOCS3 pathway. J Biochem Mol Toxicol 2021; 35:e22730. [PMID: 33522063 DOI: 10.1002/jbt.22730] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/18/2020] [Accepted: 01/20/2021] [Indexed: 12/22/2022]
Abstract
Ulcerative colitis is an autoimmune inflammatory disorder with a negative impact on the life quality of patients. Cinnamaldehyde and hesperetin were chosen due to their antioxidants and anti-inflammatory effects. This study explored the protective effects of cinnamaldehyde (40 and 90 mg/kg, po) and hesperetin (50 and 100 mg/kg, po) on 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced ulcerative colitis in rats. Cinnamaldehyde and hesperetin significantly improved macroscopic and histopathological examinations with a significant reduction in myeloperoxidase and intracellular adhesion molecule-1 expression. They significantly reduced colon oxidative stress by a significant elevation in both reduced glutathione content and superoxide dismutase activity with a significant reduction of NO content. Furthermore, cinnamaldehyde and hesperetin alleviated the inflammatory injury by a significant reduction in interleukin-6 along with suppression of nuclear factor-κB, receptor for advanced glycation end products, and tumor necrosis factor-α expression. Moreover, cinnamaldehyde and hesperetin significantly decreased p-JAK2 and p-STAT3 while significantly increased suppressors of cytokine signaling 3 (SOCS3) protein expression. In conclusion, cinnamaldehyde and hesperetin counteracted TNBS-induced ulcerative colitis through antioxidant, anti-inflammatory properties as well as modulation of the JAk2/STAT3/SOCS3 pathway.
Collapse
Affiliation(s)
| | - Eglal A Abdelaleem
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Amal A Zaki
- Department of Biochemistry, Animal Health Research Institute, Giza, Egypt
| | - Wafaa R Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
16
|
Ou-Yang H, Wu SC, Sung LY, Yang SH, Yang SH, Chong KY, Chen CM. STAT3 Is an Upstream Regulator of Granzyme G in the Maternal-To-Zygotic Transition of Mouse Embryos. Int J Mol Sci 2021; 22:ijms22010460. [PMID: 33466434 PMCID: PMC7796490 DOI: 10.3390/ijms22010460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/19/2020] [Accepted: 12/31/2020] [Indexed: 12/24/2022] Open
Abstract
The maternal-to-zygotic transition (MZT), which controls maternal signaling to synthesize zygotic gene products, promotes the preimplantation development of mouse zygotes to the two-cell stage. Our previous study reported that mouse granzyme g (Gzmg), a serine-type protease, is required for the MZT. In this study, we further identified the maternal factors that regulate the Gzmg promoter activity in the zygote to the two-cell stage of mouse embryos. A full-length Gzmg promoter from mouse genomic DNA, FL-pGzmg (−1696~+28 nt), was cloned, and four deletion constructs of this Gzmg promoter, Δ1-pGzmg (−1369~+28 nt), Δ2-pGzmg (−939~+28 nt), Δ3-pGzmg (−711~+28 nt) and Δ4-pGzmg (−417~+28 nt), were subsequently generated. Different-sized Gzmg promoters were used to perform promoter assays of mouse zygotes and two-cell stage embryos. The results showed that Δ4-pGzmg promoted the highest expression level of the enhanced green fluorescent protein (EGFP) reporter in the zygotes and two-cell embryos. The data suggested that time-specific transcription factors upregulated Gzmg by binding cis-elements in the −417~+28-nt Gzmg promoter region. According to the results of the promoter assay, the transcription factor binding sites were predicted and analyzed with the JASPAR database, and two transcription factors, signal transducer and activator of transcription 3 (STAT3) and GA-binding protein alpha (GABPα), were identified. Furthermore, STAT3 and GABPα are expressed and located in zygote pronuclei and two-cell nuclei were confirmed by immunofluorescence staining; however, only STAT3 was recruited to the mouse zygote pronuclei and two-cell nuclei injected with the Δ4-pGzmg reporter construct. These data indicated that STAT3 is a maternal transcription factor and may upregulate Gzmg to promote the MZT. Furthermore, treatment with a STAT3 inhibitor, S3I-201, caused mouse embryonic arrest at the zygote and two-cell stages. These results suggest that STAT3, a maternal protein, is a critical transcription factor and regulates Gzmg transcription activity in preimplantation mouse embryos. It plays an important role in the maternal-to-zygotic transition during early embryonic development.
Collapse
Affiliation(s)
- Huan Ou-Yang
- Department of Life Sciences, and Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; (H.O.-Y.); (S.-H.Y.)
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan;
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan;
| | - Shinn-Chih Wu
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan;
| | - Li-Ying Sung
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan;
| | - Shiao-Hsuan Yang
- Department of Life Sciences, and Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; (H.O.-Y.); (S.-H.Y.)
- Reproductive Medicine Center, Department of Gynecology, Changhua Christian Hospital, Changhua 515, Taiwan
| | - Shang-Hsun Yang
- Department of Physiology, National Cheng Kung University, Tainan 70101, Taiwan;
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 70101, Taiwan
| | - Kowit-Yu Chong
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences, and Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; (H.O.-Y.); (S.-H.Y.)
- The iEGG and Animal Biotechnology Center, and Rong-Hsing Translational Medicine Research Center, National Chung Hsing University, Taichung 402, Taiwan
- Correspondence: ; Tel.: +886-4-22856309
| |
Collapse
|
17
|
Wang N, Wang S, Wang X, Zheng Y, Yang B, Zhang J, Pan B, Gao J, Wang Z. Research trends in pharmacological modulation of tumor-associated macrophages. Clin Transl Med 2021; 11:e288. [PMID: 33463063 PMCID: PMC7805405 DOI: 10.1002/ctm2.288] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
As one of the most abundant immune cell populations in the tumor microenvironment (TME), tumor-associated macrophages (TAMs) play important roles in multiple solid malignancies, including breast cancer, prostate cancer, liver cancer, lung cancer, ovarian cancer, gastric cancer, pancreatic cancer, and colorectal cancer. TAMs could contribute to carcinogenesis, neoangiogenesis, immune-suppressive TME remodeling, cancer chemoresistance, recurrence, and metastasis. Therefore, reprogramming of the immune-suppressive TAMs by pharmacological approaches has attracted considerable research attention in recent years. In this review, the promising pharmaceutical targets, as well as the existing modulatory strategies of TAMs were summarized. The chemokine-chemokine receptor signaling, tyrosine kinase receptor signaling, metabolic signaling, and exosomal signaling have been highlighted in determining the biological functions of TAMs. Besides, both preclinical research and clinical trials have suggested the chemokine-chemokine receptor blockers, tyrosine kinase inhibitors, bisphosphonates, as well as the exosomal or nanoparticle-based targeting delivery systems as the promising pharmacological approaches for TAMs deletion or reprogramming. Lastly, the combined therapies of TAMs-targeting strategies with traditional treatments or immunotherapies as well as the exosome-like nanovesicles for cancer therapy are prospected.
Collapse
Affiliation(s)
- Neng Wang
- The Research Center for Integrative MedicineSchool of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Shengqi Wang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Xuan Wang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Yifeng Zheng
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Bowen Yang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Juping Zhang
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Bo Pan
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| | - Jianli Gao
- Academy of Traditional Chinese MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Zhiyu Wang
- The Research Center for Integrative MedicineSchool of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- The Research Center of Integrative Cancer MedicineDiscipline of Integrated Chinese and Western MedicineThe Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangdong Provincial Hospital of Chinese MedicineGuangdong Provincial Academy of Chinese Medical SciencesGuangzhouGuangdongChina
| |
Collapse
|
18
|
Jamalzadeh S, Pujari AN, Cullen PJ. A Rab escort protein regulates the MAPK pathway that controls filamentous growth in yeast. Sci Rep 2020; 10:22184. [PMID: 33335117 PMCID: PMC7746766 DOI: 10.1038/s41598-020-78470-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
MAPK pathways regulate different responses yet can share common components. Although core regulators of MAPK pathways are well known, new pathway regulators continue to be identified. Overexpression screens can uncover new roles for genes in biological processes and are well suited to identify essential genes that cannot be evaluated by gene deletion analysis. In this study, a genome-wide screen was performed to identify genes that, when overexpressed, induce a reporter (FUS1-HIS3) that responds to ERK-type pathways (Mating and filamentous growth or fMAPK) but not p38-type pathways (HOG) in yeast. Approximately 4500 plasmids overexpressing individual yeast genes were introduced into strains containing the reporter by high-throughput transformation. Candidate genes were identified by measuring growth as a readout of reporter activity. Fourteen genes were identified and validated by re-testing: two were metabolic controls (HIS3, ATR1), five had established roles in regulating ERK-type pathways (STE4, STE7, BMH1, BMH2, MIG2) and seven represent potentially new regulators of MAPK signaling (RRN6, CIN5, MRS6, KAR2, TFA1, RSC3, RGT2). MRS6 encodes a Rab escort protein and effector of the TOR pathway that plays a role in nutrient signaling. MRS6 overexpression stimulated invasive growth and phosphorylation of the ERK-type fMAPK, Kss1. Overexpression of MRS6 reduced the osmotolerance of cells and phosphorylation of the p38/HOG MAPK, Hog1. Mrs6 interacted with the PAK kinase Ste20 and MAPKK Ste7 by two-hybrid analysis. Based on these results, Mrs6 may selectively propagate an ERK-dependent signal. Identifying new regulators of MAPK pathways may provide new insights into signal integration among core cellular processes and the execution of pathway-specific responses.
Collapse
Affiliation(s)
- Sheida Jamalzadeh
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Atindra N Pujari
- Department of Biological Sciences, State University of New York at Buffalo, 532 Cooke Hall, Buffalo, NY, 14260-1300, USA
| | - Paul J Cullen
- Department of Biological Sciences, State University of New York at Buffalo, 532 Cooke Hall, Buffalo, NY, 14260-1300, USA.
| |
Collapse
|
19
|
Fang Y, Ren R, Shi H, Huang L, Lenahan C, Lu Q, Tang L, Huang Y, Tang J, Zhang J, Zhang JH. Pituitary Adenylate Cyclase-Activating Polypeptide: A Promising Neuroprotective Peptide in Stroke. Aging Dis 2020; 11:1496-1512. [PMID: 33269103 PMCID: PMC7673855 DOI: 10.14336/ad.2020.0626] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/26/2020] [Indexed: 12/11/2022] Open
Abstract
The search for viable, effective treatments for acute stroke continues to be a global priority due to the high mortality and morbidity. Current therapeutic treatments have limited effects, making the search for new treatments imperative. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a well-established cytoprotective neuropeptide that participates in diverse neural physiological and pathological activities, such as neuronal proliferation, differentiation, and migration, as well as neuroprotection. It is considered a promising treatment in numerous neurological diseases. Thus, PACAP bears potential as a new therapeutic strategy for stroke treatment. Herein, we provide an overview pertaining to the current knowledge of PACAP, its receptors, and its potential neuroprotective role in the setting of stroke, as well as various mechanisms of neuroprotection involving ionic homeostasis, excitotoxicity, cell edema, oxidative stress, inflammation, and cell death, as well as the route of PACAP administration.
Collapse
Affiliation(s)
- Yuanjian Fang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Reng Ren
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hui Shi
- 2Department of Neurosurgery, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Lei Huang
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Cameron Lenahan
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA.,5Burrell College of Osteopathic Medicine, Las Cruces, NM, USA
| | - Qin Lu
- 6Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China
| | - Lihui Tang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi Huang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiping Tang
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA.,7Department of Anesthesiology, Loma Linda University, Loma Linda, CA, USA
| | - Jianmin Zhang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - John H Zhang
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA.,7Department of Anesthesiology, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
20
|
Lan J, Xie K. miR-202-3p overexpression attenuates endometriosis-like lesions by modulating YAP-dependent transcription of S100A6 in murine models. Life Sci 2020; 265:118757. [PMID: 33197444 DOI: 10.1016/j.lfs.2020.118757] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 10/27/2020] [Accepted: 11/10/2020] [Indexed: 11/19/2022]
Abstract
AIM Recent evidence has suggested the important implications of microRNAs (miRNAs) in the processes of proliferation and tissue remodeling in endometriosis (EMS). We therefore aim to determine the role of miR-202-3p in the pathophysiology of EMS and its underlying mechanisms. METHODS Experimental endometriosis was induced in ovariectomized mice implanted with a slow-release 17-β estradiol capsule. Eutopic endometrial stromal cells (euESCs) were isolated and assayed for proliferative, invasive and apoptotic properties by EdU staining, Transwell assays, and flow cytometry. The invasive and apoptotic features in the endometrium of mice with EMS in vivo were evaluated by using immunohistochemical staining and TUNEL assays. RESULTS miR-202-3p was observed to be downregulated in the endometrial tissues of EMS patients. MiR-202-3p was also found to target YAP1 which resulted in reduced euESC proliferation and invasion and increased apoptosis. YAP1 was able to phosphorylated STAT3 which consequently upregulated S100A6 to promote the proliferative and invasive abilities of euESCs. MiR-202-3p was thereby proposed to act as an inhibitor of proliferation and tissue damage in the in vivo setting of EMS, its effects however, were able to be counteracted byS100A6, which reversed the effects of miR-202-3p on tissue injury and cell proliferation. CONCLUSION Our data together evidenced that miR-202-3p targeted YAP1 to reduce STAT3-mediated S100A6 whereby preventing the progression of EMS.
Collapse
Affiliation(s)
- Jing Lan
- The Second Department of Gynecologic Oncology, Hunan Cancer Hospital (The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University), Changsha 410013, PR China
| | - Kangling Xie
- Department of Rehabilitation Medicine, Xiangya Hospital, Central South University, Changsha 410008, PR China.
| |
Collapse
|
21
|
Xu J, Jin S, Gan F, Xiong H, Mei Z, Chen Y, Yang G. Polycyclic polyprenylated acylphloroglucinols from Garcinia xanthochymus fruits exhibit antitumor effects through inhibition of the STAT3 signaling pathway. Food Funct 2020; 11:10568-10579. [PMID: 33185634 DOI: 10.1039/d0fo02535f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The fruits of Garcinia xanthochymus can be eaten raw or processed into jams, preserves and vinegar. They provide not only vitamin and protein nutrients, but also pharmacologically active compounds, among which polycyclic polyprenylated acylphloroglucinols (PPAPs) are a major class. According to the literature, PPAPs exhibited good anti-cancer effects. This study investigated the antitumor effects and the underlying mechanism of S1 (the regioisomeric mixture of xanthochymol and guttiferone E) and S2 (the regioisomeric mixture of isoxanthochymol and cycloxanthochymol) isolated from the fruits of G. xanthochymus. In an H22 allograft mouse model, S1 and S2 could suppress the liver tumor growth and phosphorylation of STAT3. Computational modeling showed that S1 and S2 could form hydrogen bonds with the SH2 domain of STAT3. In HepG2 and MCF-7 cell lines, S1 and S2 downregulated the expression of p-STAT3Tyr705. Moreover, S1 and S2 inhibited the phosphorylation of JAK2 and Src, which are the upstream kinases of STAT3, and the expression of various STAT3-regulated genes, including anti-apoptotic (Bcl-XL, Mcl-1 and survivin), proliferative (cyclin D1) and angiogenic (VEGF) genes. As a result, S1 and S2 arrested the cell cycle and induced cell apoptosis, which were proved by the activation of cleaved caspase-3 and caspase-8. These results demonstrated that S1 and S2 from G. xanthochymus exhibited antitumor effects through the inactivation of STAT3, and could be promising candidates for cancer treatment.
Collapse
Affiliation(s)
- Jing Xu
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
22
|
STAT3 transcription factor as target for anti-cancer therapy. Pharmacol Rep 2020; 72:1101-1124. [PMID: 32880101 DOI: 10.1007/s43440-020-00156-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/17/2022]
Abstract
STATs constitute a large family of transcription activators and transducers of signals that have an important role in many cell functions as regulation of proliferation and differentiation of the cell also regulation of apoptosis and angiogenesis. STAT3 as a member of that family, recently was discovered to have a vital role in progression of different types of cancers. The activation of STAT3 was observed to regulate multiple gene functions during cancer-like cell proliferation, differentiation, apoptosis, metastasis, inflammation, immunity, cell survival, and angiogenesis. The inhibition of STAT3 activation has been an important target for cancer therapy. Inhibitors of STAT3 have been used for a long time for treatment of many types of cancers like leukemia, melanoma, colon, and renal cancer. In this review article, we summarize and discuss different drugs inhibiting the action of STAT3 and used in treatment of different types of cancer.
Collapse
|
23
|
Barclay RA, Mensah GA, Cowen M, DeMarino C, Kim Y, Pinto DO, Erickson J, Kashanchi F. Extracellular Vesicle Activation of Latent HIV-1 Is Driven by EV-Associated c-Src and Cellular SRC-1 via the PI3K/AKT/mTOR Pathway. Viruses 2020; 12:E665. [PMID: 32575590 PMCID: PMC7354524 DOI: 10.3390/v12060665] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/08/2020] [Accepted: 06/17/2020] [Indexed: 12/14/2022] Open
Abstract
HIV-1 is a global health crisis that has infected more than 37 million people. Latent reservoirs throughout the body are a major hurdle when it comes to eradicating the virus. In our previous study, we found that exosomes, a type of extracellular vesicle (EV), from uninfected cells activate the transcription of HIV-1 in latent infected cells, regardless of combination antiretroviral therapy (cART). In this study, we investigated the specific mechanism behind the EV activation of latent HIV-1. We found that phosphorylated c-Src is present in EVs of various cell lines and has the ability to activate downstream proteins such as EGFR, initiating a signal cascade. EGFR is then able to activate the PI3K/AKT/mTOR pathway, resulting in the activation of STAT3 and SRC-1, culminating in the reversal of HIV-1 latency. This was verified by examining levels of HIV-1 TAR, genomic RNA and HIV-1 Gag p24 protein in cell lines and primary cells. We found that EVs containing c-Src rescued HIV-1 despite the presence of inhibitors, validating the importance of EV-associated c-Src in latent HIV-1 activation. Lastly, we discovered an increased recruitment of p300 and NF-κB in the nucleus of EV-treated infected cells. Collectively, our data suggest that EV-associated c-Src is able to activate latent HIV-1 via the PI3K/AKT/mTOR pathway and SRC-1/p300-driven chromatin remodeling. These findings could aid in designing new strategies to prevent the reactivation of latent HIV-1 in patients under cART.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Fatah Kashanchi
- Laboratory of Molecular Virology, George Mason University, Manassas, VA 20110, USA; (R.A.B.); (G.A.M.); (M.C.); (C.D.); (Y.K.); (D.O.P.); (J.E.)
| |
Collapse
|
24
|
Lee HS, Jung JI, Kim KH, Park SJ, Kim EJ. Rhus verniciflua Stokes extract suppresses migration and invasion in human gastric adenocarcinoma AGS cells. Nutr Res Pract 2020; 14:463-477. [PMID: 33029287 PMCID: PMC7520559 DOI: 10.4162/nrp.2020.14.5.463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/27/2020] [Accepted: 05/20/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND/OBJECTIVES Many studies have suggested that Rhus verniciflua Stokes (RVS) and its extract are anticancer agents. However, RVS had limited use because it contains urushiol, an allergenic toxin. By improving an existing allergen-removal extraction method, we developed a new allergen-free Rhus verniciflua Stokes extract (RVSE) with higher flavonoid content. In this study, we examined whether RVSE inhibits the ability of AGS gastric cancer cells to migrate and invade. MATERIALS/METHODS The flavonoids content of RVSE was analyzed by HPLC. The effects of RVSE on migration and invasion in AGS cells were analyzed by each assay kit. Matrix metalloproteinase (MMP)-9, plasminogen activator inhibitor-1 (PAI-1) and urokinase-type plasminogen activator (uPA) protein expression was analyzed by protein antibody array. The Phosphorylation of signal transducer and activator of transcription (STAT) 3 were assayed by Western blot analysis. RESULTS RVSE treatment with 0-100 μg/mL dose-dependently reduced the ability of AGS cells to migrate and invade. Notably, treatment with RVSE strongly inhibited the expression of MMP-9 and uPA and the phosphorylation of STAT3. In contrast, RVSE treatment dramatically increased the expression of PAI-1. These results indicate that the inhibition of MMP-9 and uPA expression and STAT3 phosphorylation and the stimulation of PAI-1 expression contributed to the decreased migration and invasion of AGS cells treated with RVSE. CONCLUSIONS These results suggest that RVSE may be used as a natural herbal agent to reduce gastric cancer metastasis.
Collapse
Affiliation(s)
- Hyun Sook Lee
- Department of Food Science & Nutrition, Dongseo University, Busan 47011, Korea
| | - Jae In Jung
- Regional Strategic Industry Innovation Center, Hallym University, Chuncheon 24252, Korea
| | | | | | - Eun Ji Kim
- Regional Strategic Industry Innovation Center, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
25
|
Li S, Wan C, Zheng R, Fan J, Dong X, Meyer CA, Liu XS. Cistrome-GO: a web server for functional enrichment analysis of transcription factor ChIP-seq peaks. Nucleic Acids Res 2020; 47:W206-W211. [PMID: 31053864 PMCID: PMC6602521 DOI: 10.1093/nar/gkz332] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/19/2019] [Accepted: 05/02/2019] [Indexed: 01/12/2023] Open
Abstract
Characterizing the ontologies of genes directly regulated by a transcription factor (TF), can help to elucidate the TF’s biological role. Previously, we developed a widely used method, BETA, to integrate TF ChIP-seq peaks with differential gene expression (DGE) data to infer direct target genes. Here, we provide Cistrome-GO, a website implementation of this method with enhanced features to conduct ontology analyses of gene regulation by TFs in human and mouse. Cistrome-GO has two working modes: solo mode for ChIP-seq peak analysis; and ensemble mode, which integrates ChIP-seq peaks with DGE data. Cistrome-GO is freely available at http://go.cistrome.org/.
Collapse
Affiliation(s)
- Shaojuan Li
- Shanghai Key Laboratory of Tuberculosis, Clinical Translational Research Center, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Changxin Wan
- Shanghai Key Laboratory of Tuberculosis, Clinical Translational Research Center, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Rongbin Zheng
- Shanghai Key Laboratory of Tuberculosis, Clinical Translational Research Center, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jingyu Fan
- Shanghai Key Laboratory of Tuberculosis, Clinical Translational Research Center, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xin Dong
- Shanghai Key Laboratory of Tuberculosis, Clinical Translational Research Center, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Clifford A Meyer
- Department of Data Sciences, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA.,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - X Shirley Liu
- Shanghai Key Laboratory of Tuberculosis, Clinical Translational Research Center, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.,Department of Data Sciences, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA.,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| |
Collapse
|
26
|
A Phytocomplex Consisting of Tropaeolum majus L. and Salvia officinalis L. Extracts Alleviates the Inflammatory Response of Dermal Fibroblasts to Bacterial Lipopolysaccharides. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8516153. [PMID: 32566105 PMCID: PMC7261326 DOI: 10.1155/2020/8516153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/14/2020] [Accepted: 03/23/2020] [Indexed: 02/07/2023]
Abstract
Background The antimicrobial activity and effects of a phytocomplex consisting of Tropaeolum flos (T) and Salviae folium (S) extracts on the cytokine levels and transcription factors on dermal fibroblast BJ exposed to bacterial lipopolysaccharides were examined. Methods In order to select the most optimal combination ratio of the two extracts for using in vitro, the physicochemical characterization of vegetal extract mixtures was performed and the antioxidant and antibacterial activities were evaluated on five different formulations of T : S, namely, 1 : 1, 1 : 2, 2 : 1, 3 : 1, and 1 : 3. The best combination of bioactive compounds with regard to antioxidant and antibacterial activities (T : S 1 : 2) was selected for in vitro evaluation of the anti-inflammatory effect. Human dermal fibroblast BJ cells were treated with two doses of the extract mixture and then exposed to bacterial lipopolysaccharides (LPS). The levels of the cytokines involved in inflammatory response, namely, interleukin- (IL-) 6, tumor necrosis factor- (TNF-) α, IL-31, and IL-33, were quantified by ELISA, and the expression of transcription factors, namely, signal transducer and activator of transcription (STAT) 3, nuclear factor kappa B (NFκB), and phosphorylated NFκB (pNFκB), were evaluated by western blot analysis. Results The results have shown that the mixture of T : S 1 : 2 exhibited significant antibacterial effects on Staphylococcus aureus ATCC 25923. LPS exposure increased the cytokine levels in BJ cells and enhanced the NFκB expression. The pretreatment of BF cells exposed to LPS with the two doses of the extract mixture markedly inhibited the increase of IL-33 and TNF-α levels and amplified the NFκB expression and its activation, especially with the high dose. The low doses of the extract reduced NFκB expression but increased its activation. Conclusions These experimental findings suggest that the mixture of T : S 1 : 2 can exert some protection against bacterial infections and inflammation induced by LPS in BJ cells being a good therapeutic option in related conditions associated with inflammation.
Collapse
|
27
|
Role of microRNAs in epidermal growth factor receptor signaling pathway in cervical cancer. Mol Biol Rep 2020; 47:4553-4568. [PMID: 32383136 DOI: 10.1007/s11033-020-05494-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 05/03/2020] [Indexed: 12/18/2022]
Abstract
Cervical cancer is one of the most common disorders in females all around the world. Similar to other types of cancer, several signaling pathways are demonstrated to be involved in the progression of this cancer including ERK/MAPK, PI3K/AKT, apoptotic signaling pathways, Wnt, and epidermal growth factor receptor (EGFR). Various microRNAs (miRNAs) and their target genes involved in cervical cancer have been extracted from the kinds of literature of Scopus, Pubmed and Google scholar databases. Regarding the targets, some of them were found to belong in EGFR signaling pathways. The regulation patterns of these miRNA are different in cervical cancer; however, their main aim is to trigger EGFR signaling to proceed with cancer. Moreover, several predicted miRNAs were found to have some interactions with the differentially expressed genes of cervical cancer which are the members of the EGFR signaling pathway by using miRWalk 3.0 (https://mirwalk.umm.uni-heidelberg.de/) and TargetScan 7.1 (https://www.targetscan.org/vert_71/). Also, the microarray data were obtained from the NCBI-Gene Expression Omnibus (GEO) datasets of cervical cancer. In the present review, we highlight the miRNAs involved in cervical cancer and the role of their targets in the EGFR signaling pathway. Furthermore, some predicted miRNAs were the candidate to target EGFR signaling pathway members differentially expressed in cervical cancer samples compared to normal samples.
Collapse
|
28
|
Gattelli A, Hynes NE, Schor IE, Vallone SA. Ret Receptor Has Distinct Alterations and Functions in Breast Cancer. J Mammary Gland Biol Neoplasia 2020; 25:13-26. [PMID: 32080788 DOI: 10.1007/s10911-020-09445-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 02/06/2020] [Indexed: 12/15/2022] Open
Abstract
Ret receptor tyrosine kinase is a proto-oncogene that participates in development of various cancers. Several independent studies have recently identified Ret as a key player in breast cancer. Although Ret overexpression and function have been under investigation, mainly in estrogen receptor positive breast cancer, a more comprehensive analysis of the impact of recurring Ret alterations in breast cancer is needed. This review consolidates the current knowledge of Ret alterations and their potential effects in breast cancer. We discuss and integrate data on Ret changes in different breast cancer subtypes and potential function in progression, as well as the participation of distinct Ret network signaling partners in these processes. We propose that it will be essential to define a shared molecular feature of tumors with alteration in Ret receptor, be this at the genetic level or via overexpression in order to design effective therapies to target the Ret pathway. Here we review experimental evidence from basic research and pre-clinical studies concentrating on Ret alterations as potential biomarkers for recurrence, and we discuss the possibility that targeting the Ret pathway might in the future become a treatment for breast cancer.
Collapse
Affiliation(s)
- Albana Gattelli
- CONICET-UBA, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Ciudad Universitaria, C1428EGA CABA, Buenos Aires, Argentina.
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Ciudad Universitaria C1428EGA CABA, Buenos Aires, Argentina.
| | - Nancy E Hynes
- Friedrich Miescher Institute for Biomedical Research (FMI), Maulbeerstrasse 66, CH-4058, Basel, Switzerland
- University of Basel, CH-4002, Basel, Switzerland
| | - Ignacio E Schor
- CONICET-UBA, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Ciudad Universitaria, C1428EGA CABA, Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), Ciudad Universitaria, C1428EGA, CABA, Argentina
| | - Sabrina A Vallone
- CONICET-UBA, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Ciudad Universitaria, C1428EGA CABA, Buenos Aires, Argentina
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Ciudad Universitaria C1428EGA CABA, Buenos Aires, Argentina
| |
Collapse
|
29
|
Antisense lncRNA LDLRAD4-AS1 promotes metastasis by decreasing the expression of LDLRAD4 and predicts a poor prognosis in colorectal cancer. Cell Death Dis 2020; 11:155. [PMID: 32111819 PMCID: PMC7048743 DOI: 10.1038/s41419-020-2338-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/07/2020] [Accepted: 02/07/2020] [Indexed: 11/30/2022]
Abstract
Long noncoding RNAs (lncRNAs) have been revealed to play critical roles in tumor initiation and progression. The antisense lncRNA LDLRAD4-AS1 is the longest lncRNA of LDLRAD4, and its expression levels, cellular localization, precise function, and mechanism in colorectal cancer (CRC) remain unknown. In this study, we observed that lncRNA LDLRAD4-AS1 was located in the nucleus of CRC cells and that lncRNA LDLRAD4-AS1 was upregulated in most CRC specimens and cell lines. Overexpression of lncRNA LDLRAD4-AS1 was correlated with poor prognosis in CRC patients. LncRNA LDLRAD4-AS1 upregulation enhanced the migration and invasion of CRC cells in vitro and facilitated CRC metastasis in vivo. Mechanistic investigations suggested that lncRNA LDLRAD4-AS1 could decrease the expression of LDLRAD4 by disrupting the stability of LDLRAD4 mRNA, resulting in epithelial-to-mesenchymal transition (EMT) through upregulating Snail, thereby promoting metastasis in CRC. Our results demonstrated a previously unrecognized LDLRAD4-AS1-LDLRAD4-Snail regulatory axis involved in epigenetic and posttranscriptional regulation that contributes to CRC progression and metastasis.
Collapse
|
30
|
AL-Eitan LN, Alghamdi MA, Tarkhan AH, Al-Qarqaz FA. Genome-Wide Tiling Array Analysis of HPV-Induced Warts Reveals Aberrant Methylation of Protein-Coding and Non-Coding Regions. Genes (Basel) 2019; 11:E34. [PMID: 31892232 PMCID: PMC7017144 DOI: 10.3390/genes11010034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/18/2019] [Accepted: 12/22/2019] [Indexed: 12/18/2022] Open
Abstract
The human papillomaviruses (HPV) are a group of double-stranded DNA viruses that exhibit an exclusive tropism for squamous epithelia. HPV can either be low- or high-risk depending on its ability to cause benign lesions or cancer, respectively. Unsurprisingly, the majority of epigenetic research has focused on the high-risk HPV types, neglecting the low-risk types in the process. Therefore, the main objective of this study is to better understand the epigenetics of wart formation by investigating the differences in methylation between HPV-induced cutaneous warts and normal skin. A number of clear and very significant differences in methylation patterns were found between cutaneous warts and normal skin. Around 55% of the top-ranking 100 differentially methylated genes in warts were protein coding, including the EXOC4, KCNU, RTN1, LGI1, IRF2, and NRG1 genes. Additionally, non-coding RNA genes, such as the AZIN1-AS1, LINC02008, and MGC27382 genes, constituted 11% of the top-ranking 100 differentially methylated genes. Warts exhibited a unique pattern of methylation that is a possible explanation for their transient nature. Since the genetics of cutaneous wart formation are not completely known, the findings of the present study could contribute to a better understanding of how HPV infection modulates host methylation to give rise to warts in the skin.
Collapse
Affiliation(s)
- Laith N. AL-Eitan
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan;
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mansour A. Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia;
| | - Amneh H. Tarkhan
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan;
| | - Firas A. Al-Qarqaz
- Department of Internal Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan;
- Division of Dermatology, Department of Internal Medicine, King Abdullah University Hospital, Jordan University of Science and Technology, Irbid 22110, Jordan
| |
Collapse
|
31
|
Mekonnen YA, Gültas M, Effa K, Hanotte O, Schmitt AO. Identification of Candidate Signature Genes and Key Regulators Associated With Trypanotolerance in the Sheko Breed. Front Genet 2019; 10:1095. [PMID: 31803229 PMCID: PMC6872528 DOI: 10.3389/fgene.2019.01095] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/11/2019] [Indexed: 12/23/2022] Open
Abstract
African animal trypanosomiasis (AAT) is caused by a protozoan parasite that affects the health of livestock. Livestock production in Ethiopia is severely hampered by AAT and various controlling measures were not successful to eradicate the disease. AAT affects the indigenous breeds in varying degrees. However, the Sheko breed shows better trypanotolerance than other breeds. The tolerance attributes of Sheko are believed to be associated with its taurine genetic background but the genetic controls of these tolerance attributes of Sheko are not well understood. In order to investigate the level of taurine background in the genome, we compare the genome of Sheko with that of 11 other African breeds. We find that Sheko has an admixed genome composed of taurine and indicine ancestries. We apply three methods: (i) The integrated haplotype score (iHS), (ii) the standardized log ratio of integrated site specific extended haplotype homozygosity between populations (Rsb), and (iii) the composite likelihood ratio (CLR) method to discover selective sweeps in the Sheko genome. We identify 99 genomic regions harboring 364 signature genes in Sheko. Out of the signature genes, 15 genes are selected based on their biological importance described in the literature. We also identify 13 overrepresented pathways and 10 master regulators in Sheko using the TRANSPATH database in the geneXplain platform. Most of the pathways are related with oxidative stress responses indicating a possible selection response against the induction of oxidative stress following trypanosomiasis infection in Sheko. Furthermore, we present for the first time the importance of master regulators involved in trypanotolerance not only for the Sheko breed but also in the context of cattle genomics. Our finding shows that the master regulator Caspase is a key protease which plays a major role for the emergence of adaptive immunity in harmony with the other master regulators. These results suggest that designing and implementing genetic intervention strategies is necessary to improve the performance of susceptible animals. Moreover, the master regulatory analysis suggests potential candidate therapeutic targets for the development of new drugs for trypanosomiasis treatment.
Collapse
Affiliation(s)
- Yonatan Ayalew Mekonnen
- Breeding Informatics Group, Department of Animal Sciences, University of Göttingen, Göttingen, Germany
| | - Mehmet Gültas
- Breeding Informatics Group, Department of Animal Sciences, University of Göttingen, Göttingen, Germany.,Center for Integrated Breeding Research (CiBreed), University of Göttingen, Göttingen, Germany
| | - Kefena Effa
- Animal Biosciences, National Program Coordinator for African Dairy Genetic Gain, International Livestock Research Institute (ILRI), Addis Ababa, Ethiopia
| | - Olivier Hanotte
- Cells, Organisms amd Molecular Genetics, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom.,LiveGene, International Livestock Research Institute (ILRI), Addis Ababa, Ethiopia
| | - Armin O Schmitt
- Breeding Informatics Group, Department of Animal Sciences, University of Göttingen, Göttingen, Germany.,Center for Integrated Breeding Research (CiBreed), University of Göttingen, Göttingen, Germany
| |
Collapse
|
32
|
MicroRNAs as Potential Biomarkers in Atherosclerosis. Int J Mol Sci 2019; 20:ijms20225547. [PMID: 31703274 PMCID: PMC6887712 DOI: 10.3390/ijms20225547] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 11/03/2019] [Accepted: 11/05/2019] [Indexed: 01/08/2023] Open
Abstract
Atherosclerosis is a complex multifactorial disease that, despite advances in lifestyle management and drug therapy, remains to be the major cause of high morbidity and mortality rates from cardiovascular diseases (CVDs) in industrialized countries. Therefore, there is a great need in reliable diagnostic/prognostic biomarkers and effective treatment alternatives to reduce its burden. It was established that microRNAs (miRNAs/miRs), a class of non-coding single-stranded RNA molecules, can regulate the expression of genes at the post-transcriptional level and, accordingly, coordinate the cellular protein expression. Thus, they are involved not only in cell-specific physiological functions but also in the cellular and molecular mechanisms of human pathologies, including atherosclerosis. MiRNAs may be significant in the dysregulation that affects endothelial integrity, the function of vascular smooth muscle and inflammatory cells, and cellular cholesterol homeostasis that drives the initiation and growth of an atherosclerotic plaque. Besides, distinct expression patterns of several miRNAs are attributed to atherosclerotic and cardiovascular patients. In this article, the evidence indicating the multiple critical roles of miRNAs and their relevant molecular mechanisms related to atherosclerosis development and progression was reviewed. Moreover, the effects of miRNAs on atherosclerosis enabled to exploit them as novel diagnostic biomarkers and therapeutic targets that may lead to better management of atherosclerosis and CVDs.
Collapse
|
33
|
Zhao T, Feng Y, Guo M, Zhang C, Wu Q, Chen J, Guo S, Liu S, Zhou Q, Wang Z, Fan W, Zhang Y, Jia H, Feng Z. Combination of attenuated
Salmonella
carrying PD‐1 siRNA with nifuroxazide for colon cancer therapy. J Cell Biochem 2019; 121:1973-1985. [DOI: 10.1002/jcb.29432] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 10/10/2019] [Indexed: 01/05/2023]
Affiliation(s)
- Tiesuo Zhao
- Institute of Precision Medicine Xinxiang Medical University Xinxiang Henan China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy Xinxiang Medical University Xinxiang Henan China
- Department of Immunology Xinxiang Medical University Xinxiang Henan China
| | - Yuchen Feng
- Institute of Precision Medicine Xinxiang Medical University Xinxiang Henan China
- Department of Immunology Xinxiang Medical University Xinxiang Henan China
- Department of Interventional Radiology The First Hospital of Handan Handan China
| | - Mengmeng Guo
- Institute of Precision Medicine Xinxiang Medical University Xinxiang Henan China
- Department of Pathology Xinxiang Medical University Xinxiang Henan China
| | - Chaohui Zhang
- Institute of Precision Medicine Xinxiang Medical University Xinxiang Henan China
| | - Qiang Wu
- Department of Pathology Xinxiang Medical University Xinxiang Henan China
| | - Jian Chen
- Institute of Precision Medicine Xinxiang Medical University Xinxiang Henan China
| | - Sheng Guo
- Institute of Precision Medicine Xinxiang Medical University Xinxiang Henan China
- Department of Immunology Xinxiang Medical University Xinxiang Henan China
| | - Shenzhen Liu
- Institute of Precision Medicine Xinxiang Medical University Xinxiang Henan China
| | - Qingsa Zhou
- Institute of Precision Medicine Xinxiang Medical University Xinxiang Henan China
| | - Zizhong Wang
- Institute of Precision Medicine Xinxiang Medical University Xinxiang Henan China
| | - Wenyan Fan
- Institute of Precision Medicine Xinxiang Medical University Xinxiang Henan China
| | - Yongxi Zhang
- Department of Oncology The Third Affiliated Hospital of Xinxiang Medical University Xinxiang Henan China
| | - Huijie Jia
- Institute of Precision Medicine Xinxiang Medical University Xinxiang Henan China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy Xinxiang Medical University Xinxiang Henan China
- Department of Pathology Xinxiang Medical University Xinxiang Henan China
| | - Zhiwei Feng
- Institute of Precision Medicine Xinxiang Medical University Xinxiang Henan China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy Xinxiang Medical University Xinxiang Henan China
- Department of Immunology Xinxiang Medical University Xinxiang Henan China
| |
Collapse
|
34
|
Li R, Wei X, Jiang DS. Protein methylation functions as the posttranslational modification switch to regulate autophagy. Cell Mol Life Sci 2019; 76:3711-3722. [PMID: 31222372 PMCID: PMC11105718 DOI: 10.1007/s00018-019-03161-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 05/10/2019] [Accepted: 05/28/2019] [Indexed: 02/07/2023]
Abstract
Studies over the past decades have elucidated the critical role of autophagy in human health and diseases. Although the processes of autophagy in the cytoplasm have been well studied, the posttranscriptional and epigenetic regulation mechanisms of autophagy are still poorly understood. Protein methylation, including histone methylation and non-histone protein methylation, is the most important type of posttranscriptional and epigenetic modification. Recent studies have shown that protein methylation is associated with effects on autophagosome formation, autophagy-related protein expression, and signaling pathway activation, but the details are still unclear. Thus, it is important to summarize the current status and discuss the future directions of research on protein methylation in the context of autophagy.
Collapse
Affiliation(s)
- Rui Li
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
- NHC Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Ding-Sheng Jiang
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.
- NHC Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China.
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
35
|
Cheng Z, Lei Z, Yang P, Si A, Xiang D, Tang X, Guo G, Zhou J, Hüser N. Exosome-transmitted p120-catenin suppresses hepatocellular carcinoma progression via STAT3 pathways. Mol Carcinog 2019; 58:1389-1399. [PMID: 30997702 DOI: 10.1002/mc.23022] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/22/2019] [Accepted: 04/07/2019] [Indexed: 12/14/2022]
Abstract
Hepatocellular carcinoma (HCC) is a fatal disease with increasing morbidity and poor prognosis due to surgical recurrence and metastasis. Moreover, the molecular mechanism of HCC progression remains unclear. Although the role of p120-catenin (p120ctn) in liver cancer is well studied, the effects of secreted p120ctn transported by exosomes are less understood. Here, we show that p120ctn in exosomes secreted from liver cancer cells suppresses HCC cell proliferation and metastasis and expansion of liver cancer stem cells (CSCs). Mechanically, exosome p120ctn inhibits HCC cell progression via the STAT3 pathway, and the STAT3 inhibitor S3I-201 abolishes the observed effects on growth, metastasis, and self-renewal ability between exosome p120ctn-treated HCC cells and control cells. Taken together, we propose that p120ctn-containing exosomes derived from cancer cells inhibit the progression of liver cancer and may offer a new therapeutic strategy.
Collapse
Affiliation(s)
- Zhangjun Cheng
- Hepato-Pancreato-Biliary Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhengqing Lei
- Hepato-Pancreato-Biliary Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Pinghua Yang
- Department of Hepatic Surgery, The Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Anfeng Si
- Department of Minimally Invasive Surgery, The Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Daimin Xiang
- National Liver Cancer Science Center, Second Military Medical University, Shanghai, China
| | - Xuewu Tang
- Hepato-Pancreato-Biliary Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Guangmeng Guo
- Hepato-Pancreato-Biliary Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jiahua Zhou
- Hepato-Pancreato-Biliary Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Norbert Hüser
- Department of Surgery, Klinikum rechts der Isar, The Technical University of Munich, Munich, Germany
| |
Collapse
|
36
|
Zhou Y, Xu S, Xia H, Gao Z, Huang R, Tang E, Jiang X. Long noncoding RNA FEZF1-AS1 in human cancers. Clin Chim Acta 2019; 497:20-26. [PMID: 31276636 DOI: 10.1016/j.cca.2019.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/27/2019] [Accepted: 07/01/2019] [Indexed: 12/24/2022]
Abstract
Long noncoding RNAs (lncRNAs) have been shown to play key roles in various human tumors. Ectopic expression of the lncRNA FEZ finger zinc 1 antisense 1 (FEZF1-AS1) have been reported in different cancers, including colorectal cancer, gastric neoplasia, hepatocellular carcinoma and so on. Summarizing all literature correlated with FEZF1-AS1, it is obvious that FEZF1-AS1 is mainly involved in tumorigenesis and progression through competing endogenous RNA (ceRNA) which sponges tumor-suppressive microRNA (miRNA) and recruiting mechanism. Moreover, the aberrant expression of FEZF1-AS1 is related to clinical features of patients with cancers, and regulates cellular proliferation, anti-apoptosis, invasion and metastasis through diverse underlying mechanisms. The role of FEZF1-AS1 in carcinogenesis and progression suggests that it may be a potential diagnostic biomarker or a novel therapeutic target for cancers.
Collapse
Affiliation(s)
- Yuanshi Zhou
- Department of HPB Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 246 Xuefu-ro, Harbin 150086, China
| | - Shuwan Xu
- Department of HPB Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 246 Xuefu-ro, Harbin 150086, China
| | - Haoming Xia
- Department of HPB Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 246 Xuefu-ro, Harbin 150086, China
| | - Zewei Gao
- Department of HPB Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 246 Xuefu-ro, Harbin 150086, China
| | - Rongju Huang
- Department of HPB Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 246 Xuefu-ro, Harbin 150086, China
| | - Enyu Tang
- Department of HPB Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 246 Xuefu-ro, Harbin 150086, China
| | - Xingming Jiang
- Department of HPB Surgery, The 2nd Affiliated Hospital of Harbin Medical University, 246 Xuefu-ro, Harbin 150086, China.
| |
Collapse
|
37
|
Zhuo D, Wu Y, Luo J, Deng L, Niu X. CSTP1 inhibits IL-6 expression through targeting Akt/FoxO3a signaling pathway in bladder cancer cells. Exp Cell Res 2019; 380:80-89. [PMID: 31002815 DOI: 10.1016/j.yexcr.2019.04.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 10/27/2022]
Abstract
CSTP1, a recently identified protein phosphotase, is frequently repressed in bladder cancers. Previous results showed that CSTP1 over-expression inhibited cell cycle progression and promoted apoptosis through dephosphorylating Akt kinase at Ser473 site in bladder cancer cells, but the mechanisms how CSTP1 exerted tumor suppressive activity remains unclear. In this study, we analyzed the gene expression profile changes that affected by CSTP1 overexpression by microarray, and reported that CSTP1 decreased IL-6 expression/secretion in bladder cancer cells and re-expression of IL-6 abrogated CSTP1's tumor suppressive activity. We also found that FoxO3a occupy IL-6 gene promoter and repressed IL mRNA transcription. Further results showed that decreased expression of IL-6 in CSTP1-overexpressing cells inactivated Stat3 transcriptional factor, which resulted in the down-regulation of cyclin D1, Bcl-xl expression. Spearman correlation analysis revealed that the mRNA level of CSTP1 correlated inversely with that of IL-6 in bladder cancer tissues. In conclusion, our studies revealed that protein phosphotase CSTP1 inhibited IL-6 expression through targeting Akt/FoxO3a signaling pathway and IL-6 inactivated Stat3 was necessary for CSTP1's tumor suppressive function.
Collapse
Affiliation(s)
- Dexiang Zhuo
- The Central Laboratory of Sanming First Hospital Affiliated to Fujian Medical University, Sanming City, 365000, China
| | - Yongyang Wu
- The Central Laboratory of Sanming First Hospital Affiliated to Fujian Medical University, Sanming City, 365000, China
| | - Jia Luo
- The Central Laboratory of Sanming First Hospital Affiliated to Fujian Medical University, Sanming City, 365000, China
| | - Ling Deng
- Department of Clinical Medical Oncology, Qingyuan People's Hospital, The Six Affiliated Hosptial of Guangzhou Medical University, Qingyuan City, 511518, China
| | - Xiaohua Niu
- Department of Gastrointestinal Surgery, Qingyuan People's Hospital, The Six Affiliated Hosptial of Guangzhou Medical University, Qingyuan City, 511518, China.
| |
Collapse
|
38
|
Lu YC, Wang P, Wang J, Ma R, Lee SC. PCNA and JNK1-Stat3 pathways respectively promotes and inhibits diabetes-associated centrosome amplification by targeting at the ROCK1/14-3-3σ complex in human colon cancer HCT116 cells. J Cell Physiol 2018; 234:11511-11523. [PMID: 30478982 PMCID: PMC6587713 DOI: 10.1002/jcp.27813] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/01/2018] [Indexed: 12/13/2022]
Abstract
We have recently reported that type 2 diabetes promotes centrosome amplification via enhancing the expression, biding, and centrosome translocation of rho‐associated coiled‐coil containing protein kinase 1 (ROCK1)/14‐3‐3σ complex in HCT116 cells. In the functional proteomic study, we further investigated the molecular pathways underlying the centrosome amplification using HCT116 cells. We found that treatment of HCT116 cells with high glucose, insulin, and palmitic acid triggered the centrosome amplification and increased the expressions of proliferating cell nuclear antigen (PCNA), nucleophosmin (NPM), and 14‐3‐3σ. Individual knockdown of PCNA, NPM, or 14‐3‐3σ inhibited the centrosome amplification. Knockdown of PCNA inhibited the treatment‐increased expression of ROCK1, whereas knockdown of ROCK1 did not affect the PCNA expression. High glucose, insulin, and palmitic acid also increased the expressions of c‐Jun N‐terminal kinase‐1 (JNK1) and signal transducer and activator of transcription 3 (Stat3), individual knockdown of which upregulated the treatment‐increased expression of 14‐3‐3σ and promoted the centrosome amplification. In contrast, overexpression of JNK1 inhibited the centrosome amplification. Knockdown of Stat3 enhanced the centrosome translocation of 14‐3‐3σ. Moreover, we showed that knockdown of JNK1 inhibited the treatment‐increased expression of Stat3. Knockdown of PCNA, JNK1, or Stat3 did not have an effect on NPM and vice versa. In conclusion, our results suggest that PCNA and JNK1‐Stat3 pathways respectively promotes and feedback inhibits the centrosome amplification by targeting at the ROCK1/14‐3‐3σ complex, and NPM serves as an independent signal for the centrosome amplification.
Collapse
Affiliation(s)
- Yu Cheng Lu
- School of Life Sciences, Shanxi University, Taiyuan, Shanxi, China.,Central Laboratory, Linyi People's Hospital, Linyi, Shandong, China
| | - Pu Wang
- School of Life Sciences, Shanxi University, Taiyuan, Shanxi, China
| | - Jie Wang
- School of Acupuncture and Moxibustion, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, China
| | - Ronald Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Shao Chin Lee
- School of Life Sciences, Shanxi University, Taiyuan, Shanxi, China.,School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, China
| |
Collapse
|
39
|
Huang H, Bu Y, Zhang X, Liu J, Zhu L, Fang Y. LINC01433 promotes hepatocellular carcinoma progression via modulating the miR‐1301/STAT3 axis. J Cell Physiol 2018; 234:6116-6124. [PMID: 30317567 DOI: 10.1002/jcp.27366] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 08/14/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Haijin Huang
- Department of General Surgery Hongze District People's Hospital Hongze China
| | - Yan‐Zhi Bu
- Department of General Surgery Lianshui County People's Hospital Huai'an China
| | - Xiao‐Yu Zhang
- Division of Gastrointestinal Surgery, Department of General Surgery Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Juan Liu
- Operating Room, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Li‐Yao Zhu
- Department of Hepatology The Fourth People's Hospital of Huai'an Huai'an China
| | - Yong Fang
- Department of General Surgery Jiangwan Hospital of Shanghai Hongkou District, No. 22 Changzhong Road Hongkou District Shanghai China
| |
Collapse
|
40
|
Long J, Yin Y, Guo H, Li S, Sun Y, Zeng C, Zhu W. The mechanisms and clinical significance of PDCD4 in colorectal cancer. Gene 2018; 680:59-64. [PMID: 30243936 DOI: 10.1016/j.gene.2018.09.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 09/17/2018] [Accepted: 09/19/2018] [Indexed: 12/14/2022]
Abstract
In recent years, the incidence and mortality of colorectal cancer (CRC) have been on a global upward trend. There is an urgent need for effective tools to prevent and treat CRC and reduce morbidity and mortality of CRC patients. Recent evidence suggests that programmed cell death 4 (PDCD4), a novel tumor suppressor gene, inhibits tumor progression at transcriptional and translational levels and regulates multiple signal transduction pathways. However, little is known about the precise mechanisms regulating PDCD4 expression in CRC. In addition, several studies have demonstrated that the expression of in CRC is down-regulated or even absent. PDCD4 is therefore considered to be an independent prognostic factor in CRC and may be a potential support diagnostic tool for distinguishing in normal colon tissue, benign adenoma and CRC. This review will focus on the expression of PDCD4 in CRC and the relevant molecular mechanisms.
Collapse
Affiliation(s)
- Jiali Long
- Department of Pathology, School of Basic Medicine, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Yuting Yin
- Department of Pathology, School of Basic Medicine, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Haina Guo
- Department of Pathology, Dongguan Maternal and Child Health Hospital, Dongguan 523013, Guangdong Province, China
| | - Shuling Li
- Department of Pathology, Dongguan Hospital of Southern Medical University, Dongguan 523059, Guangdong Province, China
| | - Yanqin Sun
- Department of Pathology, School of Basic Medicine, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Chao Zeng
- Department of Pathology, School of Basic Medicine, Guangdong Medical University, Dongguan 523808, Guangdong Province, China.
| | - Wei Zhu
- Department of Pathology, School of Basic Medicine, Guangdong Medical University, Dongguan 523808, Guangdong Province, China.
| |
Collapse
|
41
|
Qureshi AA, Khan DA, Mushtaq S, Ye SQ, Xiong M, Qureshi N. δ-Tocotrienol feeding modulates gene expression of EIF2, mTOR, protein ubiquitination through multiple-signaling pathways in chronic hepatitis C patients. Lipids Health Dis 2018; 17:167. [PMID: 30031388 PMCID: PMC6054847 DOI: 10.1186/s12944-018-0804-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/26/2018] [Indexed: 12/12/2022] Open
Abstract
Background δ-Tocotrienol is a naturally occurring proteasome inhibitor, which has the capacity to inhibit proliferation and induce apoptosis in several cancer cells obtained from several organs of humans, and other cancer cell lines. Moreover, results of plasma total mRNAs after δ-tocotrienol feeding to hepatitis C patients revealed significant inhibition in the expression of pro-inflammatory cytokines (TNF-α, VCAM1, proteasome subunits) and induction in the expression of ICAM1 and IFN-γ after post-treatment. This down-regulation of proteasome subunits leads to autophagy, apoptosis of immune cells and several genes. The present study describes RNA-sequence analysis of plasma total mRNAs obtained from δ-tocotrienol treatment of hepatitis C patients on gene expression regulated by proteasome. Methods Pooled specimens of plasma total mRNAs of pre-dose versus post-dose of δ-tocotrienol treatment of hepatitis C patients were submitted to RNA-sequence analyses. The data based on > 1 and 8-fold expression changes of 2136 genes were uploaded into “Ingenuity Pathway Analyses (IPA)” for core analysis, which describes possible canonical pathways, upstream regulators, diseases and functional metabolic networks. Results The IPA of “molecules” indicated fold change in gene expression of 953 molecules, which covered several categories of biological biomarkers. Out of these, gene expression of 220 related to present study, 12 were up-regulated, and 208 down-regulated after δ-tocotrienol treatment. The gene expression of transcription regulators (ceramide synthase 3 and Mohawk homeobox) were up-regulated, and gene expression of 208 molecules were down-regulated, involved in several biological functions (HSP90AB1, PSMC3, CYB5R4, NDUFB1, CYP2R1, TNFRF1B, VEGFA, GPR65, PIAS1, SFPQ, GPS2, EIF3F, GTPBP8, EIF4A1, HSPA14, TLR8, TUSSC2). IPA of “causal network” indicated gene regulators (676), in which 76 down-regulated (26 s proteasomes, interleukin cytokines, and PPAR-ligand-PPA-Retinoic acid-RXRα, PPARγ-ligand-PPARγ-Retinoic acid-RARα, IL-21, IL-23) with significant P-values. The IPA of “diseases and functions” regulators (85) were involved with cAMP, STAT2, 26S proteasome, CSF1, IFNγ, LDL, TGFA, and microRNA-155-5p, miR-223, miR-21-5p. The IPA of “upstream analysis” (934) showed 57 up-regulated (mainly 38 microRNAs) and 64 gene regulators were down-regulated (IL-2, IL-5, IL-6, IL-12, IL-13, IL-15, IL-17, IL-18, IL-21, IL-24, IL-27, IL-32), interferon β-1a, interferon γ, TNF-α, STAT2, NOX1, prostaglandin J2, NF-κB, 1κB, TCF3, and also miRNA-15, miRNA-124, miRNA-218-5P with significant activation of Z-Score (P < 0.05). Conclusions This is first report describing RNA-sequence analysis of δ-tocotrienol treated plasma total mRNAs obtained from chronic hepatitis C patients, that acts via multiple-signaling pathways without any side-effects. These studies may lead to development of novel classes of drugs for treatment of chronic hepatitis C patients. Electronic supplementary material The online version of this article (10.1186/s12944-018-0804-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Asaf A Qureshi
- Department of Biomedical Science, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO, 64108, USA.
| | - Dilshad A Khan
- Department of Chemical Pathology and Endocrinology, Armed Forces Institute of Pathology (AFIP), National University of Medical Sciences, Rawalpindi, 64000, Pakistan
| | - Shahida Mushtaq
- Department of Chemical Pathology and Endocrinology, Armed Forces Institute of Pathology (AFIP), National University of Medical Sciences, Rawalpindi, 64000, Pakistan
| | - Shui Qing Ye
- Department of Biomedical Science, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO, 64108, USA.,Division of Experimental and Translational Genetics, Department of Pediatrics, Childern's Mercy Hospital, 2401 Gillham Road, Kansas City, MO, 64108, USA.,Department of Biomedical and Health Informatics, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO, 64108, USA
| | - Min Xiong
- Department of Biomedical Science, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO, 64108, USA.,Division of Experimental and Translational Genetics, Department of Pediatrics, Childern's Mercy Hospital, 2401 Gillham Road, Kansas City, MO, 64108, USA
| | - Nilofer Qureshi
- Department of Biomedical Science, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO, 64108, USA.,Pharmacology/Toxicology, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO, 64108, USA
| |
Collapse
|
42
|
Yu X, Lin XJ, Wang S, Liu X, Li W, Kou BX, Chai M, Chen D, Liu X, Wang X. Antitumor Efficacy of Huqizhengxiao (HQZX) Decoction Based on Inhibition of Telomerase Activity in Nude Mice of Hepatocarcinoma Xenograft. Integr Cancer Ther 2018; 17:1216-1224. [PMID: 29978739 PMCID: PMC6247564 DOI: 10.1177/1534735418785999] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Objective: Huqizhengxiao (HQZX) decoction is a mixture of
traditional Chinese medicines comprising 10 herbs, with inhibitory effects on
hepatocarcinoma. The aim of the study is to observe the antitumor efficacy and
mechanism of HQZX decoction in nude mice with hepatocellular carcinoma
xenografts. Methods: HepG2-luc subcutaneous hepatocarcinoma was
established in nude mice. The mice were divided into 5 groups: control,
cinobufagin, HQZXS, HQZXM, and HQZXH with doses 13.52, 27.03, and 54.06 g/kg,
respectively. HQZX decoction was prepared for intraperitoneal intragastric
administration for 3 weeks. Tumor growth was measured with Vernier calipers and
in vivo imaging system. α-Fetoprotein (AFP) was determined by radioimmunoassay.
Tumor necrosis factor–α (TNF-α) was measured with enzyme-linked immunosorbent
assay (ELISA) assay. Telomerase activity was measured with polymerase chain
reaction–ELISA. Nuclear mitosis and necrosis were observed with
hematoxylin-eosin stain. Apoptotic proteins of caspase-3, Bcl-2, and Bax were
examined by Western blot. Signaling molecules of ERK, mTOR, and STAT3 were
measured with Luminex assay. Results: HQZX decoction showed good
inhibition of HepG2-luc xenografts. Compared with control group, the relative
tumor proliferation rate was less than 60% in the HQZXH and HQZXS. The tumor
inhibition rate of HQZXH group reached 52% ± 15%. Relative average optical
density values of the HQZXS and HQZXH groups decreased significantly. The
mitotic index in HQZXS, HQZXM, and HQZXH groups decreased greatly. Telomerase
activity of HQZXS was clearly reduced, and, the caspase-3 expression upregulated
in HQZXH group. Bcl-2 expression was downregulated in HQZXS and HQZXH. The
ratios of p-ERK/ERK and p-STAT3/STAT3 in HQZXS group were significantly
downregulated. Conclusion: HQZX decoction can clearly inhibit the
growth of hepatocellular carcinoma and induce tumor apoptosis. Its antitumor
mechanism may be related to reducing telomerase activity and regulating the
STAT3 and ERK signal pathway.
Collapse
Affiliation(s)
- XiaoXiao Yu
- 1 Beijing You-An Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xue-Jun Lin
- 1 Beijing You-An Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Shuang Wang
- 1 Beijing You-An Hospital, Capital Medical University, Beijing, People's Republic of China
| | - XiuHong Liu
- 1 Beijing You-An Hospital, Capital Medical University, Beijing, People's Republic of China.,2 Beijing Institute of Hepatology, Beijing, People's Republic of China
| | - WeiHua Li
- 1 Beijing You-An Hospital, Capital Medical University, Beijing, People's Republic of China.,2 Beijing Institute of Hepatology, Beijing, People's Republic of China
| | - Bu-Xin Kou
- 1 Beijing You-An Hospital, Capital Medical University, Beijing, People's Republic of China.,2 Beijing Institute of Hepatology, Beijing, People's Republic of China
| | - MengYin Chai
- 1 Beijing You-An Hospital, Capital Medical University, Beijing, People's Republic of China.,2 Beijing Institute of Hepatology, Beijing, People's Republic of China
| | - DeXi Chen
- 1 Beijing You-An Hospital, Capital Medical University, Beijing, People's Republic of China.,2 Beijing Institute of Hepatology, Beijing, People's Republic of China
| | - XiaoNi Liu
- 1 Beijing You-An Hospital, Capital Medical University, Beijing, People's Republic of China.,2 Beijing Institute of Hepatology, Beijing, People's Republic of China
| | - XiaoJun Wang
- 1 Beijing You-An Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
43
|
Gene Profile Expression Related to Type I Interferons in HT-29 Cells Exposed to Cryptosporidium parvum. Jundishapur J Microbiol 2018. [DOI: 10.5812/jjm.63071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
44
|
Lim RZL, Li L, Yong EL, Chew N. STAT-3 regulation of CXCR4 is necessary for the prenylflavonoid Icaritin to enhance mesenchymal stem cell proliferation, migration and osteogenic differentiation. Biochim Biophys Acta Gen Subj 2018; 1862:1680-1692. [PMID: 29679717 DOI: 10.1016/j.bbagen.2018.04.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 03/22/2018] [Accepted: 04/16/2018] [Indexed: 01/07/2023]
Abstract
Mesenchymal stem cell (MSC) dysfunction has been implicated in the pathogenesis of osteoporosis. MSCs derived from osteoporotic subjects demonstrate significant impairment in proliferation, adhesion and chemotaxis, and osteogenic differentiation, leading to reduced functional bone-forming osteoblasts and ultimately nett bone loss and osteoporosis. Epimedium herbs and its active compound Icaritin (ICT) have been used in Chinese ethnopharmacology for the treatment of metabolic bone diseases. Using an in-vitro cell culture model, we investigated the benefits of ICT treatment in enhancing MSC proliferation, migration and osteogenic differentiation, and provide novel data to describe its mechanism of action. ICT enhances MSC proliferation, chemotaxis to stromal cell-derived factor-1 (SDF-1) and osteogenic differentiation through the activation of signal transduction activator transcription factor 3 (STAT-3), with a consequential up-regulation in the expression and activity of cysteine (C)-X-C motif chemokine receptor 4 (CXCR4). These findings provide a strong basis for future clinical studies to confirm the therapeutic potential of ICT for the prevention and treatment of osteoporosis and fragility fractures.
Collapse
Affiliation(s)
- R Z L Lim
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - L Li
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - E L Yong
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - N Chew
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Division of Infectious Diseases, National University Hospital, Singapore.
| |
Collapse
|
45
|
Mizoguchi A, Yano A, Himuro H, Ezaki Y, Sadanaga T, Mizoguchi E. Clinical importance of IL-22 cascade in IBD. J Gastroenterol 2018; 53:465-474. [PMID: 29075900 PMCID: PMC5866830 DOI: 10.1007/s00535-017-1401-7] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/04/2017] [Indexed: 02/07/2023]
Abstract
IL-22 is a relatively new cytokine that is characterized by several unique biological properties. In the intestines, the effect of IL-22 is restricted mainly to non-lymphoid cells such as epithelial cells. Interestingly, the expression pattern and major cellular source of IL-22 have distinct difference between large and small intestines. IL-22 possesses an ability to constitutively activate STAT3 for promoting epithelial cell regeneration and reinforcing mucosal barrier integrity through stimulating the expression of anti-bacterial peptide and mucins. Of note, IL-22 is characterized as a two-faced cytokine that can play not only protective but also deleterious roles in the intestinal inflammation depending on the cytokine environment such as the expression levels of IL-23, T-bet, and IL-22 binding protein. Most importantly, clinical relevance of IL-22 to inflammatory bowel disease has been well highlighted. Mucosal healing, which represents the current therapeutic goal for IBD, can be induced by IL-22. Indeed, indigo naturalis, which can activate IL-22 pathway through Ahr, has been shown in a clinical trial to exhibit a strong therapeutic effect on ulcerative colitis. Despite the beneficial effect of IL-22, continuous activation of the IL-22 pathway increases the risk of colitis-associated cancer, particularly in patients with an extended history of IBD. This review article discusses how IL-22 regulates colitis, how beneficial versus deleterious effects of IL-22 is determined, and why IL-22 represents a promising target for IBD therapy.
Collapse
Affiliation(s)
- Atsushi Mizoguchi
- Department of Immunology, Kurume University School of Medicine, Asahi-machi, Kurume, Fukuoka, 830-0011, Japan.
- IBD Center, Kurume University Hospital, Kurume, Japan.
| | - Arisa Yano
- Department of Immunology, Kurume University School of Medicine, Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Hidetomo Himuro
- Department of Immunology, Kurume University School of Medicine, Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Yui Ezaki
- Department of Immunology, Kurume University School of Medicine, Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Takayuki Sadanaga
- Department of Immunology, Kurume University School of Medicine, Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Emiko Mizoguchi
- Department of Immunology, Kurume University School of Medicine, Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
- IBD Center, Kurume University Hospital, Kurume, Japan
| |
Collapse
|
46
|
Weyrich A, Jeschek M, Schrapers KT, Lenz D, Chung TH, Rübensam K, Yasar S, Schneemann M, Ortmann S, Jewgenow K, Fickel J. Diet changes alter paternally inherited epigenetic pattern in male Wild guinea pigs. ENVIRONMENTAL EPIGENETICS 2018; 4:dvy011. [PMID: 29992049 PMCID: PMC6031029 DOI: 10.1093/eep/dvy011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/19/2018] [Accepted: 04/06/2018] [Indexed: 05/14/2023]
Abstract
Epigenetic modifications, of which DNA methylation is the most stable, are a mechanism conveying environmental information to subsequent generations via parental germ lines. The paternal contribution to adaptive processes in the offspring might be crucial, but has been widely neglected in comparison to the maternal one. To address the paternal impact on the offspring's adaptability to changes in diet composition, we investigated if low protein diet (LPD) in F0 males caused epigenetic alterations in their subsequently sired sons. We therefore fed F0 male Wild guinea pigs with a diet lowered in protein content (LPD) and investigated DNA methylation in sons sired before and after their father's LPD treatment in both, liver and testis tissues. Our results point to a 'heritable epigenetic response' of the sons to the fathers' dietary change. Because we detected methylation changes also in the testis tissue, they are likely to be transmitted to the F2 generation. Gene-network analyses of differentially methylated genes in liver identified main metabolic pathways indicating a metabolic reprogramming ('metabolic shift'). Epigenetic mechanisms, allowing an immediate and inherited adaptation may thus be important for the survival of species in the context of a persistently changing environment, such as climate change.
Collapse
Affiliation(s)
- A Weyrich
- Leibniz-Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Street 17, D-10315 Berlin, Germany
| | - M Jeschek
- Leibniz-Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Street 17, D-10315 Berlin, Germany
- Berlin Center for Genomics in Biodiversity Research, Koenigin-Luise-Street 6-8, 14195 Berlin, Germany
| | - K T Schrapers
- Leibniz-Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Street 17, D-10315 Berlin, Germany
| | - D Lenz
- Leibniz-Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Street 17, D-10315 Berlin, Germany
| | - T H Chung
- Zymoresearch, EpiQuest, 17062 Murphy Avenue, Irvine, CA 92614, USA
| | - K Rübensam
- Leibniz-Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Street 17, D-10315 Berlin, Germany
| | - S Yasar
- Leibniz-Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Street 17, D-10315 Berlin, Germany
| | - M Schneemann
- Leibniz-Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Street 17, D-10315 Berlin, Germany
| | - S Ortmann
- Leibniz-Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Street 17, D-10315 Berlin, Germany
| | - K Jewgenow
- Leibniz-Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Street 17, D-10315 Berlin, Germany
| | - J Fickel
- Leibniz-Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Street 17, D-10315 Berlin, Germany
- Institute for Biochemistry and Biology, Potsdam University, Karl-Liebknecht-Street 24-25, 14476 Potsdam, Germany
| |
Collapse
|
47
|
Rivnyak A, Kiss P, Tamas A, Balogh D, Reglodi D. Review on PACAP-Induced Transcriptomic and Proteomic Changes in Neuronal Development and Repair. Int J Mol Sci 2018; 19:ijms19041020. [PMID: 29596316 PMCID: PMC5979407 DOI: 10.3390/ijms19041020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/21/2018] [Accepted: 03/26/2018] [Indexed: 12/19/2022] Open
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a neuropeptide with widespread occurrence and diverse biological effects. Among its several different effects, of special importance is the action of PACAP on neuronal proliferation, differentiation and migration, and neuroprotection. The neuroprotective mechanism of PACAP is both direct and indirect, via neuronal and non-neuronal cells. Several research groups have performed transcriptomic and proteomic analysis on PACAP-mediated genes and proteins. Hundreds of proteins have been described as being involved in the PACAP-mediated neuroprotection. In the present review we summarize the few currently available transcriptomic data potentially leading to the proteomic changes in neuronal development and protection. Proteomic studies focusing on the neuroprotective role of PACAP are also reviewed and discussed in light of the most intriguing and promising effect of this neuropeptide, which may possibly have future therapeutic potential.
Collapse
Affiliation(s)
- Adam Rivnyak
- Department of Anatomy, MTA-PTE PACAP Research Team, Neuroscience Centre, University of Pecs Medical School, 7624 Pécs, Hungary.
| | - Peter Kiss
- Department of Anatomy, MTA-PTE PACAP Research Team, Neuroscience Centre, University of Pecs Medical School, 7624 Pécs, Hungary.
| | - Andrea Tamas
- Department of Anatomy, MTA-PTE PACAP Research Team, Neuroscience Centre, University of Pecs Medical School, 7624 Pécs, Hungary.
| | - Dorottya Balogh
- Department of Anatomy, MTA-PTE PACAP Research Team, Neuroscience Centre, University of Pecs Medical School, 7624 Pécs, Hungary.
| | - Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Team, Neuroscience Centre, University of Pecs Medical School, 7624 Pécs, Hungary.
| |
Collapse
|
48
|
Li Y, Zhang W, Zuo Y, Zhu T, Pang Y, Li T, Li Q. Label-Free Quantitative Proteomic Reveals Differentially Expressed Proteins in Aeromonas-Immunostimulated Leukocytes of Lampetra japonica. Curr Microbiol 2018. [DOI: 10.1007/s00284-018-1468-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
49
|
Huang C, Zhang Z, Chen L, Lee HW, Ayrapetov MK, Zhao TC, Hao Y, Gao J, Yang C, Mehta GU, Zhuang Z, Zhang X, Hu G, Chin YE. Acetylation within the N- and C-Terminal Domains of Src Regulates Distinct Roles of STAT3-Mediated Tumorigenesis. Cancer Res 2018. [PMID: 29531159 DOI: 10.1158/0008-5472.can-17-2314] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Chao Huang
- Translation Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
- Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhe Zhang
- Department of Urology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lihan Chen
- Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hank W Lee
- Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Marina K Ayrapetov
- Departments of Surgery and Medicine, Brown University School of Medicine-Rhode Island Hospital, Providence, Rhode Island
| | - Ting C Zhao
- Departments of Surgery and Medicine, Brown University School of Medicine-Rhode Island Hospital, Providence, Rhode Island
| | - Yimei Hao
- Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jinsong Gao
- Departments of Surgery and Medicine, Brown University School of Medicine-Rhode Island Hospital, Providence, Rhode Island
| | - Chunzhang Yang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland
| | - Gautam U Mehta
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland
| | - Zhengping Zhuang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland
| | - Xiaoren Zhang
- Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guohong Hu
- Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Y Eugene Chin
- Translation Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
- Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
50
|
Tian F, Jia L, Chu Z, Han H, Zhang Y, Cai J. MicroRNA-519a inhibits the proliferation and promotes the apoptosis of ovarian cancer cells through targeting signal transducer and activator of transcription 3. Exp Ther Med 2018; 15:1819-1824. [PMID: 29434770 PMCID: PMC5776609 DOI: 10.3892/etm.2017.5600] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 09/12/2017] [Indexed: 01/01/2023] Open
Abstract
Ovarian cancer is a highly prevalent cancer among women. Recent studies have indicated that microRNAs (miRs) may serve important roles in the pathogenesis of ovarian cancer. miR-519a was observed to be downregulated in tissue samples of patients with ovarian cancer; however, its role in ovarian cancer requires further investigation. The aim of the present study was to examine the role of miR-519a in the pathogenesis of ovarian cancer and determine its direct target. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was performed to examine the expression of miR-519a in 20 patients ovarian cancer and 20 normal ovarian tissue samples. Subsequently, SKOV3 cells were cultured and transfected with miR-519a mimics, while MTT and Annexin V assays were performed to investigate the role of miR-519a in the proliferation and apoptosis of SKOV3 cells. In addition, RT-qPCR and western blotting were used to determine the expression levels of miR-519a, signal transducer and activator of transcription 3 (STAT3), myeloid cell leukemia 1 (Mcl-1) and B-cell lymphoma-extra large (Bcl-xl) in untransfected and miR-519a mimic-transfected SKOV3 cells. Dual-luciferase reporter assay was also performed to confirm whether STAT3 was a direct target of miR-519a. The results revealed that miR-519a was significantly downregulated in tissue samples of patients with ovarian cancer as compared with the normal ovarian tissues. Furthermore, transient overexpression of miR-519a inhibited the proliferation and promoted the apoptosis of SKOV3 cells, as well as decreased the mRNA and protein expression levels of STAT3, Mcl-1 and Bcl-xl. Finally, dual-luciferase reporter assay confirmed that STAT3 was a direct target of miR-519a. In conclusion, the present study proved for the first time that miR-519a functions as a tumor suppressor by targeting STAT3 in ovarian cancer, suggesting that miR-519a may be a potential biomarker for the diagnosis and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Fei Tian
- Obstetrics and Gynecology Teaching and Research Section, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
- Department of Gynecology, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Ligang Jia
- Department of Gynecology, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Zhaoping Chu
- Department of Gynecology, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Hua Han
- Department of Gynecology, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Yuan Zhang
- Department of Gynecology, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Jianhui Cai
- Surgery Teaching and Research Section, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
- Department of Surgery, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|